1
|
Miura T, Kouzu H, Tanno M, Tatekoshi Y, Kuno A. Role of AMP deaminase in diabetic cardiomyopathy. Mol Cell Biochem 2024; 479:3195-3211. [PMID: 38386218 DOI: 10.1007/s11010-024-04951-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/24/2024] [Indexed: 02/23/2024]
Abstract
Diabetes mellitus is one of the major causes of ischemic and nonischemic heart failure. While hypertension and coronary artery disease are frequent comorbidities in patients with diabetes, cardiac contractile dysfunction and remodeling occur in diabetic patients even without comorbidities, which is referred to as diabetic cardiomyopathy. Investigations in recent decades have demonstrated that the production of reactive oxygen species (ROS), impaired handling of intracellular Ca2+, and alterations in energy metabolism are involved in the development of diabetic cardiomyopathy. AMP deaminase (AMPD) directly regulates adenine nucleotide metabolism and energy transfer by adenylate kinase and indirectly modulates xanthine oxidoreductase-mediated pathways and AMP-activated protein kinase-mediated signaling. Upregulation of AMPD in diabetic hearts was first reported more than 30 years ago, and subsequent studies showed similar upregulation in the liver and skeletal muscle. Evidence for the roles of AMPD in diabetes-induced fatty liver, sarcopenia, and heart failure has been accumulating. A series of our recent studies showed that AMPD localizes in the mitochondria-associated endoplasmic reticulum membrane as well as the sarcoplasmic reticulum and cytosol and participates in the regulation of mitochondrial Ca2+ and suggested that upregulated AMPD contributes to contractile dysfunction in diabetic cardiomyopathy via increased generation of ROS, adenine nucleotide depletion, and impaired mitochondrial respiration. The detrimental effects of AMPD were manifested at times of increased cardiac workload by pressure loading. In this review, we briefly summarize the expression and functions of AMPD in the heart and discuss the roles of AMPD in diabetic cardiomyopathy, mainly focusing on contractile dysfunction caused by this disorder.
Collapse
Affiliation(s)
- Tetsuji Miura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 15-4-1, Maeda-7, Teine-Ku, Sapporo, 006-8585, Japan.
| | - Hidemichi Kouzu
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masaya Tanno
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Nursing, Sapporo Medical University School of Health Sciences, Sapporo, Japan
| | - Yuki Tatekoshi
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Atsushi Kuno
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
2
|
Ishii T, Seya N, Taguri M, Wakui H, Yoshimura A, Tamura K. Allopurinol, Febuxostat, and Nonuse of Xanthine Oxidoreductase Inhibitor Treatment in Patients Receiving Hemodialysis: A Longitudinal Analysis. Kidney Med 2024; 6:100896. [PMID: 39347518 PMCID: PMC11437761 DOI: 10.1016/j.xkme.2024.100896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024] Open
Abstract
Rationale & Objective Allopurinol and febuxostat, which are xanthine oxidoreductase inhibitors, have been widely used as uric acid-lowering medications. However, evidence regarding their cardiovascular effects in hemodialysis is insufficient. This study compared the effects of allopurinol and febuxostat on mortality and cardiovascular outcomes in patients receiving hemodialysis. Study Design A retrospective observational cohort study. Setting & Participants Data of 6,791 patients who had no history of topiroxostat usage and underwent maintenance hemodialysis between March 2016 and March 2019 at Yokohama Daiichi Hospital, Zenjinkai, and its affiliated dialysis clinics in Japan's Kanagawa and Tokyo metropolitan areas were collected. Exposure Allopurinol, febuxostat, and nontreatment. Outcomes All-cause mortality, cardiovascular disease (CVD) events, heart failure (HF), acute myocardial infarction (AMI), and stroke. Analytical Approach For the main analyses, marginal structural Cox proportional hazards models were used to estimate HRs adjusted for time-varying confounding and selection bias because of censoring. Results Allopurinol and febuxostat showed significantly better survival than nontreatment for all-cause mortality (HR, 0.40; 95% CI, 0.30-0.54 and HR, 0.49; 95% CI, 0.38-0.63, respectively), without significant difference between allopurinol and febuxostat. Allopurinol showed significantly better survival than nontreatment, whereas febuxostat did not for CVD events (HR, 0.89; 95% CI, 0.84-0.95 and HR, 1.01; 95% CI, 0.96-1.07, respectively), HF (HR, 0.71; 95% CI, 0.56-0.90 and HR, 1.03; 95% CI, 0.87-1.21, respectively), and AMI (HR, 0.48; 95% CI, 0.25-0.91 and HR, 0.76; 95% CI, 0.49-1.19, respectively). No comparisons showed significant results for stroke. Limitations The ratio of renal or intestinal excretion of uric acid and uremic toxins could not be elucidated, and we could not investigate gene polymorphism because of the large number of cases. Conclusions Allopurinol and febuxostat improved survival for all-cause mortality. Allopurinol and not febuxostat reduced the risk of CVD events, HF, and AMI.
Collapse
Affiliation(s)
- Takeo Ishii
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Nephrology, Yokohama Daiichi Hospital Zenjinkai, Yokohama, Japan
| | - Nodoka Seya
- Department of Health Data Science, Tokyo Medical University, Tokyo, Japan
| | - Masataka Taguri
- Department of Health Data Science, Tokyo Medical University, Tokyo, Japan
| | - Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ashio Yoshimura
- Department of Health Data Science, Tokyo Medical University, Tokyo, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Nephrology and Hypertension, Yokohama City University Medical Center, Yokohama, Japan
| |
Collapse
|
3
|
Annesi L, Tossetta G, Borghi C, Piani F. The Role of Xanthine Oxidase in Pregnancy Complications: A Systematic Review. Antioxidants (Basel) 2024; 13:1234. [PMID: 39456486 PMCID: PMC11505381 DOI: 10.3390/antiox13101234] [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: 08/22/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Xanthine oxidoreductase (XOR) is an enzyme involved in the oxidation of hypoxanthine and xanthine to uric acid. XOR has two isoforms: xanthine dehydrogenase and xanthine oxidase (XO). XO plays a major role in oxidative stress, causing the formation of reactive oxygen species. In the present study, we aimed to summarize the evidence on the association between XO and pregnancy complications. The PRISMA checklist guided the reporting of the data. We conducted systematic searches in the PubMed and Web of Science databases to identify all human studies investigating XO in pregnancy diseases up to June 2024. A total of 195 references have been identified and 14 studies were included. Most studies focused on women with PE and GD. Overall, all the included studies found a statistically significant increase in maternal, placental, and/or fetal XO levels, activity, or tissue expression in women with pregnancy complications, compared to those with uncomplicated pregnancies. Although promising, the quality and dimension of the included studies do not allow for a definitive answer to the question of whether XO may play a crucial role in pregnancy complications. Future studies are warranted to confirm if XO could represent a prognostic and therapeutic marker in pregnancy complications and their impact on long-term maternal and offspring cardiovascular health.
Collapse
Affiliation(s)
- Lorenzo Annesi
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (L.A.); (C.B.)
| | - Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy;
| | - Claudio Borghi
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (L.A.); (C.B.)
| | - Federica Piani
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (L.A.); (C.B.)
| |
Collapse
|
4
|
Hoang TNN, Ho LBN, Nguyen KNN, Dong TAD, Le THA. The inhibition activity on tyrosinase, xanthine oxidase, and lipase of Musa balbisiana parts grown in Vietnam. Food Sci Nutr 2024; 12:7428-7437. [PMID: 39479626 PMCID: PMC11521665 DOI: 10.1002/fsn3.4364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 11/02/2024] Open
Abstract
Musa balbisiana Colla plant belongs to the Musaceae family, which is well-known for its nutritional and pharmacological properties. This study aimed to evaluate the anti-xanthine oxidase, anti-lipase, and anti-tyrosinase activities of samples of corm, pseudostem, inflorescence, fruit, peel, and seed of M. balbisiana. The results show that seed performed the highest capacity in the inhibition of three investigated enzymes, with IC50 values of 290.25 μg/mL (anti-tyrosinase), 141.51 μg/mL (anti-xanthine oxidase), and 25.66 μg/mL (anti-lipase). In addition, while fruit showed better performance in anti-lipase inhibition (IC50: 32.72 μg/mL), corm pointed out higher capability on anti-xanthine oxidase (IC50: 160.67 μg/mL) and anti-tyrosinase (IC50: 334.43 μg/mL). The data show the high potential of the application of M. balbisiana parts in medical aesthetics, dietary supplements, or medicine. This is the first report to systematically present the biological activities of all parts of M. balbisiana grown in Vietnam.
Collapse
Affiliation(s)
- Thi Ngoc Nhon Hoang
- Food Science and Technology FacultyHo Chi Minh City University of Industry and Trade (HUIT)Ho Chi Minh CityVietnam
| | - Le Bao Ngoc Ho
- Food Science and Technology FacultyHo Chi Minh City University of Industry and Trade (HUIT)Ho Chi Minh CityVietnam
| | - Kim Nhat Nguyet Nguyen
- Food Science and Technology FacultyHo Chi Minh City University of Industry and Trade (HUIT)Ho Chi Minh CityVietnam
| | - Thi Anh Dao Dong
- Department of Food Technology, Chemical Engineering FacultyHo Chi Minh City University of Technology (HCMUT)Ho Chi Minh CityVietnam
- Vietnam National University Ho Chi Minh CityHo Chi Minh CityVietnam
| | - Thi Hong Anh Le
- Food Science and Technology FacultyHo Chi Minh City University of Industry and Trade (HUIT)Ho Chi Minh CityVietnam
| |
Collapse
|
5
|
Ali N, Taher A, Islam N, Sarna NZ, Islam F. Evaluation of the relationship between xanthine oxidase activity and metabolic syndrome in a population group in Bangladesh. Sci Rep 2024; 14:20380. [PMID: 39223331 PMCID: PMC11369145 DOI: 10.1038/s41598-024-71733-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024] Open
Abstract
Xanthine oxidase (XO) is an enzyme that converts hypoxanthine into xanthine and xanthine into uric acid, which is then eliminated by the kidneys. Serum XO has been linked to diabetes, hypertension, liver dysfunction, and cardiovascular diseases. However, limited information exists on the relationship between serum XO activity and MetS. This study aimed to analyze the relationship between XO activity and metabolic syndrome (MetS) and its components in an adult population group in Bangladesh A total of 601 participants aged ≥18 years were included in the study. MetS was defined based on the criteria set by the National Cholesterol Education Program-Adult Treatment Panel III (NCEP-ATP III). Serum XO activity was measured using the enzyme-linked immunosorbent assay (ELISA), while other biochemical parameters were measured using colorimetric methods. The relationship between serum XO and MetS levels was determined through multivariate logistic regression analysis. Serum XO activity was found to be significantly higher in females (6.17 ± 3.77 U/L) as compared to males (4.00 ± 2.77 U/L) (p < 0.001). Furthermore, participants with MetS had significantly higher mean levels of serum XO (5.34 ± 3.39 U/L) than those without MetS (3.86 ± 2.90 U/L) (p < 0.001). The prevalence of MetS and its components, such as blood pressure and blood glucose increased across the XO quartiles (p < 0.001). Regression analysis indicated that XO activity was significantly and independently associated with the prevalence of MetS (at least p < 0.05 for all cases) and its components, including elevated blood pressure, high blood glucose, and low HDL-C (at least p < 0.05 for all cases). In conclusion, individuals with MetS had significantly higher XO levels than those without MetS. Serum XO activity showed an independent association with MetS and some of its components. Therefore, XO might serve as a useful marker of MetS. Prospective studies are needed to determine the underlying mechanisms linking XO and MetS.
Collapse
Affiliation(s)
- Nurshad Ali
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh.
| | - Abu Taher
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Nayeemul Islam
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Nusrat Zaman Sarna
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Farjana Islam
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| |
Collapse
|
6
|
Li X, Chen D, Qi C, Yang Y, Guo K, Ma C, Tian J, Li J, Zhang L, Wang B, Xiao Z, Ye F. Identification of a novel xanthine oxidoreductase inhibitor for hyperuricemia treatment with high efficacy and safety profile. Biomed Pharmacother 2024; 178:117223. [PMID: 39094541 DOI: 10.1016/j.biopha.2024.117223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024] Open
Abstract
Hyperuricemia is with growing incidence and of high risk to develop into gout and other metabolic diseases. The key enzyme catalyzing uric acid synthesis, xanthine oxidoreductase (XOR) is a vital target for anti-hyperuricemic drugs, while XOR inhibitors characterized as both potent and safe are currently in urgent need. In this study, a novel small molecule compound, CC15009, was identified as a specific XOR inhibitor. CC15009 exerted strongest in vitro XOR inhibitory activity among current XOR inhibitors. It also showed favorable dose-dependent uric acid-lowering effects in two different XOR substrate-induced hyperuricemic mouse models, which was significantly superior than the current first-line drug, allopurinol. Mechanically, the direct binding of CC15009 against XOR was confirmed by molecular docking and SPR analysis. The inhibition mode was competitive and reversible. Besides, the potential antioxidant activity of CC15009 was indicated by its strong inhibitory activity against the oxidized isoform of XOR, which reduced ROS generation as the byproduct. Regarding the safety concerns of current XOR inhibitors, especially in cardiovascular risks, the safety of CC15009 was comprehensively evaluated. No significant abnormality was observed in the acute, subacute toxicity tests and mini-AMES test. Notably, there was no obvious inhibition of CC15009 against cardiac ion channels, including hERG, Nav1.5, Cav1.2 at the concentration of 30 μM, indicating its lower cardiovascular risk. Taken together, our results supported CC15009 as a candidate of high efficacy and safety profile to treat hyperuricemia through direct XOR inhibition.
Collapse
Affiliation(s)
- Xuechen Li
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China; Diabetes Research Center of Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dongting Chen
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China; Diabetes Research Center of Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chufan Qi
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China; Diabetes Research Center of Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yajun Yang
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Kaijing Guo
- Department of Pharmaceutical Analysis, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Chen Ma
- Department of Pharmaceutical Analysis, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jinying Tian
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China; Diabetes Research Center of Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiang Li
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China; Diabetes Research Center of Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lu Zhang
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Baolian Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Drug Metabolism, Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD study, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Zhiyan Xiao
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Fei Ye
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China; Diabetes Research Center of Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
7
|
Özer Y, Cansever MŞ, Turan H, Bayramoğlu E, Bingöl Aydın D, İşat E, Ceyhun E, Zubarioğlu T, Aktuğlu Zeybek AÇ, Kıykım E, Evliyaoğlu O. Pteridine and tryptophan pathways in children with type 1 diabetes: Isoxanthopterin as an indicator of endothelial dysfunction. J Pharm Biomed Anal 2024; 243:116072. [PMID: 38437786 DOI: 10.1016/j.jpba.2024.116072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 03/06/2024]
Abstract
AIM Type 1 diabetes (T1D) and its complications are known to be associated with oxidative stress. Pteridine derivatives and indoleamine 2,3-dioxygenase (IDO) activity can be used as biomarkers in the evaluation of oxidative stress. In this study, our aim is to compare the concentrations of serum and urinary pteridine derivatives, as well as serum IDO activity, in children and adolescents diagnosed with T1D and those in a healthy control group. METHOD A cross-sectional study was performed and included 93 patients with T1D and 71 healthy children. Serum and urine biopterin, neopterin, monapterin, pterin, isoxanthopterin, and pterin-6-carboxylic acid (6PTC) and serum tryptophan and kynurenine levels were analyzed and compared with healthy controls. High-performance liquid chromatography was used for the analysis of pteridine derivatives, tryptophan, and kynurenine. Xanthine oxidase (XO) activity, a marker of oxidative stress, was defined by measurement of serum and urine isoxanthopterin. As an indicator of indolamine 2,3-dioxygenase (IDO) activity, the ratio of serum kynurenine/tryptophan was used. RESULTS Serum isoxanthopterin and tryptophan concentrations were increased, and serum 6PTC concentration was decreased in children with T1D (p=0.01, p=0.021, p<0.001, respectively). In children with T1D, IDO activity was not different from healthy controls (p>0.05). Serum neopterin level and duration of diabetes were weakly correlated (p=0.045, r=0.209); urine neopterin/creatinine and isoxanthopterin/creatinine levels were weakly correlated with HbA1c levels (p=0.005, r=0.305; p=0.021, r=0.249, respectively). Urine pterin/creatinine level negatively correlated with body mass index-SDS. (p=0.015, r=-0.208). CONCLUSION We found for the first time that isoxanthopterin levels increased and 6PTC levels decreased in children and adolescents with T1D. Elevated isoxanthopterin levels suggest that the XO activity is increased in TID. Increased XO activity may be an indicator of vascular complications reflecting T1D-related endothelial dysfunction.
Collapse
Affiliation(s)
- Yavuz Özer
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Department of Pediatric Endocrinology, Istanbul, Turkey.
| | - Mehmet Şerif Cansever
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Department Metabolic Diseases and Nutrition, Istanbul, Turkey
| | - Hande Turan
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Department of Pediatric Endocrinology, Istanbul, Turkey
| | - Elvan Bayramoğlu
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Department of Pediatric Endocrinology, Istanbul, Turkey
| | - Dilek Bingöl Aydın
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Department of Pediatric Endocrinology, Istanbul, Turkey
| | - Esra İşat
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Department Metabolic Diseases and Nutrition, Istanbul, Turkey
| | - Emre Ceyhun
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Department of Pediatrics, Istanbul, Turkey
| | - Tanyel Zubarioğlu
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Department Metabolic Diseases and Nutrition, Istanbul, Turkey
| | - Ayşe Çiğdem Aktuğlu Zeybek
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Department Metabolic Diseases and Nutrition, Istanbul, Turkey
| | - Ertuğrul Kıykım
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Department Metabolic Diseases and Nutrition, Istanbul, Turkey
| | - Olcay Evliyaoğlu
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Department of Pediatric Endocrinology, Istanbul, Turkey
| |
Collapse
|
8
|
Kataoka N, Imamura T. Clinical implication of hyperuricemia on the recurrence of atrial fibrillation after catheter ablation. J Arrhythm 2024; 40:651. [PMID: 38939766 PMCID: PMC11199839 DOI: 10.1002/joa3.13047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 06/29/2024] Open
Affiliation(s)
- Naoya Kataoka
- Second Department of Internal MedicineUniversity of ToyamaToyamaJapan
| | - Teruhiko Imamura
- Second Department of Internal MedicineUniversity of ToyamaToyamaJapan
| |
Collapse
|
9
|
Zhang M, Perng W, Rifas-Shiman SL, Aris IM, Oken E, Hivert MF. Metabolomic signatures for blood pressure from early to late adolescence: findings from a U.S. cohort. Metabolomics 2024; 20:52. [PMID: 38722414 PMCID: PMC11195684 DOI: 10.1007/s11306-024-02110-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/19/2024] [Indexed: 05/18/2024]
Abstract
INTRODUCTION Metabolite signatures for blood pressure (BP) may reveal biomarkers, elucidate pathogenesis, and provide prevention targets for high BP. Knowledge regarding metabolites associated with BP in adolescence remains limited. OBJECTIVES Investigate the associations between metabolites and adolescent BP, both cross-sectionally (in early and late adolescence) and prospectively (from early to late adolescence). METHODS Participants are from the Project Viva prospective cohort. During the early (median: 12.8 years; N = 556) and late (median: 17.4 years; N = 501) adolescence visits, we conducted untargeted plasma metabolomic profiling and measured systolic (SBP) and diastolic BP (DBP). We used linear regression to identify metabolites cross-sectionally associated with BP at each time point, and to assess prospective associations of changes in metabolite levels from early to late adolescence with late adolescence BP. We used Weighted Gene Correlation Network Analysis and Spearman's partial correlation to identify metabolite clusters associated with BP at each time point. RESULTS In the linear models, higher androgenic steroid levels were consistently associated with higher SBP and DBP in early and late adolescence. A cluster of 59 metabolites, mainly composed of androgenic steroids, correlated with higher SBP and DBP in early adolescence. A cluster primarily composed of fatty acid lipids was marginally associated with higher SBP in females in late adolescence. Multiple metabolites, including those in the creatine and purine metabolism sub-pathways, were associated with higher SBP and DBP both cross-sectionally and prospectively. CONCLUSION Our results shed light on the potential metabolic processes and pathophysiology underlying high BP in adolescents.
Collapse
Affiliation(s)
- Mingyu Zhang
- Division of General Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, CO-1309, #204, Boston, MA, 02215, USA.
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA.
| | - Wei Perng
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Lifecourse Epidemiology of Adiposity & Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sheryl L Rifas-Shiman
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Izzuddin M Aris
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Emily Oken
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Marie-France Hivert
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
10
|
Kang J, Li Y, Lee S, Yu K, Cho J. Pioglitazone-induced alterations of purine metabolism in healthy male subjects. Clin Transl Sci 2024; 17:e13834. [PMID: 38771175 PMCID: PMC11107522 DOI: 10.1111/cts.13834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024] Open
Abstract
Pioglitazone is class of thiazolidinediones that activates peroxisome proliferator-activated receptors (PPARs) in adipocytes to improve glucose metabolism and insulin sensitivity and has been used as a treatment for type 2 diabetes. However, the underlying mechanisms of associated pioglitazone-induced effects remain unclear. Our study aimed to investigate endogenous metabolite alterations associated with pioglitazone administration in healthy male subjects using an untargeted metabolomics approach. All subjects received 30 mg of pioglitazone once daily in the assigned sequence and period. Urine samples were collected before pioglitazone administration and for 24 h after 7 days of administration. A total of 1465 compounds were detected and filtered using a coefficient of variance below 30% and 108 metabolites were significantly altered upon pioglitazone administration via multivariate statistical analysis. Fourteen significant metabolites were identified using authentic standards and public libraries. Additionally, pathway analysis revealed that metabolites from purine and beta-alanine metabolisms were significantly altered after pioglitazone administration. Further analysis of quantification of metabolites from purine metabolism, revealed that the xanthine/hypoxanthine and uric acid/xanthine ratios were significantly decreased at post-dose. Pioglitazone-dependent endogenous metabolites and metabolic ratio indicated the potential effect of pioglitazone on the activation of PPAR and fatty acid synthesis. Additional studies involving patients are required to validate these findings.
Collapse
Affiliation(s)
- Jihyun Kang
- Department of Clinical Pharmacology and TherapeuticsSeoul National University College of Medicine and HospitalSeoulKorea
- Kidney Research Institute, Seoul National University Medical Research CenterSeoulKorea
| | - Yufei Li
- Department of Clinical Pharmacology and TherapeuticsSeoul National University College of Medicine and HospitalSeoulKorea
| | - SeungHwan Lee
- Department of Clinical Pharmacology and TherapeuticsSeoul National University College of Medicine and HospitalSeoulKorea
| | - Kyung‐Sang Yu
- Department of Clinical Pharmacology and TherapeuticsSeoul National University College of Medicine and HospitalSeoulKorea
- Department of Biomedical SciencesSeoul National University College of MedicineSeoulKorea
| | - Joo‐Youn Cho
- Department of Clinical Pharmacology and TherapeuticsSeoul National University College of Medicine and HospitalSeoulKorea
- Kidney Research Institute, Seoul National University Medical Research CenterSeoulKorea
- Department of Biomedical SciencesSeoul National University College of MedicineSeoulKorea
| |
Collapse
|
11
|
Wen S, Arakawa H, Tamai I. Uric acid in health and disease: From physiological functions to pathogenic mechanisms. Pharmacol Ther 2024; 256:108615. [PMID: 38382882 DOI: 10.1016/j.pharmthera.2024.108615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/02/2024] [Accepted: 02/17/2024] [Indexed: 02/23/2024]
Abstract
Owing to renal reabsorption and the loss of uricase activity, uric acid (UA) is strictly maintained at a higher physiological level in humans than in other mammals, which provides a survival advantage during evolution but increases susceptibility to certain diseases such as gout. Although monosodium urate (MSU) crystal precipitation has been detected in different tissues of patients as a trigger for disease, the pathological role of soluble UA remains controversial due to the lack of causality in the clinical setting. Abnormal elevation or reduction of UA levels has been linked to some of pathological status, also known as U-shaped association, implying that the physiological levels of UA regulated by multiple enzymes and transporters are crucial for the maintenance of health. In addition, the protective potential of UA has also been proposed in aging and some diseases. Therefore, the role of UA as a double-edged sword in humans is determined by its physiological or non-physiological levels. In this review, we summarize biosynthesis, membrane transport, and physiological functions of UA. Then, we discuss the pathological involvement of hyperuricemia and hypouricemia as well as the underlying mechanisms by which UA at abnormal levels regulates the onset and progression of diseases. Finally, pharmacological strategies for urate-lowering therapy (ULT) are introduced, and current challenges in UA study and future perspectives are also described.
Collapse
Affiliation(s)
- Shijie Wen
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Arakawa
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Ikumi Tamai
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
12
|
Bolognesi A, Bortolotti M, Battelli MG, Polito L. Gender Influence on XOR Activities and Related Pathologies: A Narrative Review. Antioxidants (Basel) 2024; 13:211. [PMID: 38397809 PMCID: PMC10885918 DOI: 10.3390/antiox13020211] [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: 12/21/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Taking into account the patient's gender is the first step towards more precise and egalitarian medicine. The gender-related divergences observed in purine catabolism and their pathological consequences are good examples of gender medicine differences. Uric acid is produced by the activity of xanthine oxidoreductase (XOR). The serum levels of both XOR activity and uric acid differ physiologically between the genders, being higher in men than in women. Their higher levels have been associated with gout and hypertension, as well as with vascular, cardiac, renal, and metabolic diseases. The present review analyzes the gender-related differences in these pathological conditions in relation to increases in the serum levels of XOR and/or uric acid and the opportunity for gender-driven pharmacological treatment.
Collapse
Affiliation(s)
| | | | - Maria Giulia Battelli
- Department of Medical and Surgical Sciences—DIMEC, Alma Mater Studiorum, University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy; (M.B.); (L.P.)
| | | |
Collapse
|
13
|
Mudgal R, Singh S. Xanthine Oxidoreductase in the Pathogenesis of Endothelial Dysfunction: An Update. Curr Hypertens Rev 2024; 20:10-22. [PMID: 38318826 DOI: 10.2174/0115734021277772240124075120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/04/2023] [Accepted: 12/19/2023] [Indexed: 02/07/2024]
Abstract
Xanthine oxidoreductase (XOR) is a rate-limiting enzyme in the formation of uric acid (UA) and is involved in the generation of reactive oxygen species (ROS). Overproduction of ROS has been linked to the pathogenesis of hypertension, atherosclerosis, and cardiovascular disease, with multiple studies over the last 30 years demonstrating that XOR inhibition is beneficial. The involvement of XOR and its constituents in the advancement of chronic inflammation and ROS, which are responsible for endothelial dysfunction, is the focus of this evidence-based review. An overabundance of XOR products and ROS appears to drive the inflammatory response, resulting in significant endothelium damage. It has also been demonstrated that XOR activity and ED are connected. Diabetes, hypertension, and cardiovascular disease are all associated with endothelial dysfunction. ROS mainly modifies the activity of vascular cells and can be important in normal vascular physiology as well as the development of vascular disease. Suppressing XOR activity appears to decrease endothelial dysfunction, probably because it lessens the generation of reactive oxygen species and the oxidative stress brought on by XOR. Although there has long been a link between higher vascular XOR activity and worse clinical outcomes, new research suggests a different picture in which positive results are mediated by XOR enzymatic activity. Here in this study, we aimed to review the association between XOR and vascular endothelial dysfunction. The prevention and treatment approaches against vascular endothelial dysfunction in atherosclerotic disease.
Collapse
Affiliation(s)
- Rajat Mudgal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India
| | - Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Bihar, India
| |
Collapse
|
14
|
Zhang Y, Ding X, Guo L, Zhong Y, Xie J, Xu Y, Li H, Zheng D. Comprehensive analysis of the relationship between xanthine oxidoreductase activity and chronic kidney disease. iScience 2023; 26:107332. [PMID: 37927553 PMCID: PMC10622700 DOI: 10.1016/j.isci.2023.107332] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/19/2023] [Accepted: 07/05/2023] [Indexed: 11/07/2023] Open
Abstract
Chronic kidney disease (CKD) is a common disease that seriously endangers human health. However, the potential relationship between xanthine oxidoreductase (XOR) activity and CKD remains unclear. In this study, we used clinical data, CKD datasets from the Gene Expression Omnibus database, and untargeted metabolomics to explain the relationship between XOR activity and CKD. First, XOR activity showed high correlation with the biomarkers of CKD, such as serum creatinine, blood urea nitrogen, uric acid, and estimated glomerular filtration rate. Then, we used least absolute shrinkage and selection operator logical regression algorithm and random forest algorithm to screen CKD molecular markers from differentially expressed genes, and the results of qRT-PCR of XDH, KOX-1, and ROMO1 were in accordance with the results of bioinformatics analyses. In addition, untargeted metabolomics analysis revealed that the purine metabolism pathway was significantly enriched in CKD patients in the simulated models of kidney fibrosis.
Collapse
Affiliation(s)
- Yiyuan Zhang
- Department of Nephrology, The Affiliated Huai’an Hospital of Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, China
| | - Xiaobao Ding
- Department of Nephrology, The Affiliated Huai’an Hospital of Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, China
- Department of Pharmacology, Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Lihao Guo
- Department of Nephrology, The Affiliated Huai’an Hospital of Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, China
| | - Yanan Zhong
- Department of Nephrology, The Affiliated Huai’an Hospital of Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, China
| | - Juan Xie
- Department of Nephrology, The Affiliated Huai’an Hospital of Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, China
| | - Yong Xu
- Department of Nephrology, The Affiliated Huai’an Hospital of Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, China
| | - Hailun Li
- Department of Nephrology, The Affiliated Huai’an Hospital of Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, China
| | - Donghui Zheng
- Department of Nephrology, The Affiliated Huai’an Hospital of Xuzhou Medical University and Huai’an Second People’s Hospital, Huai’an, China
| |
Collapse
|
15
|
Singh A, Singh K, Sharma A, Kaur K, Chadha R, Singh Bedi PM. Past, present and future of xanthine oxidase inhibitors: design strategies, structural and pharmacological insights, patents and clinical trials. RSC Med Chem 2023; 14:2155-2191. [PMID: 37974965 PMCID: PMC10650961 DOI: 10.1039/d3md00316g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/06/2023] [Indexed: 11/19/2023] Open
Abstract
Xanthine oxidase, a molybdo-flavoenzyme, and an isoform of xanthine dehydrogenase both exist as xanthine oxidoreductase and are responsible for purine catabolism. Xanthine oxidase is more involved in pathological conditions when extensively modulated. Elevation of xanthine oxidase is not only the prime cause of gout but is also responsible for various hyperuricemia associated pathological conditions like diabetes, chronic wounds, cardiovascular disorders, Alzheimer's disease, etc. Currently available xanthine oxidase inhibitors in clinical practice (allopurinol, febuxostat and topiroxostat) suffer from fatal side effects that pose a serious problem to the healthcare system, raising global emergency to develop novel, potent and safer xanthine oxidase inhibitors. This review will provide key and systematic information about: a. design strategies (inspired from both marketed drugs in clinical practice and natural products), structural insights and pharmacological output (xanthine oxidase inhibition and associated activities) of various pre-clinical candidates reported by various research groups across the globe in the past two decades; b. patented xanthine oxidase inhibitors published in the last three decades and c. clinical trials and their outcomes on approved drug candidates. Information generated in this review has suggested fragment-based drug design (FBDD) and molecular hybridization techniques to be most suitable for development of desired xanthine oxidase inhibitors as one provides high selectivity toward the enzyme and the other imparts multifunctional properties to the structure and both may possess capabilities to surpass the limitations of currently available clinical drugs. All in combination will exclusively update researchers working on xanthine oxidase inhibitors and allied areas and potentially help in designing rational, novel, potent and safer xanthine oxidase inhibitors that can effectively tackle xanthine oxidase related disease conditions and disorders.
Collapse
Affiliation(s)
- Atamjit Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Karanvir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Aman Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Kirandeep Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
| | - Renu Chadha
- University Institute of Pharmaceutical Sciences, Panjab University Chandigarh 160014 India
| | - Preet Mohinder Singh Bedi
- Department of Pharmaceutical Sciences, Guru Nanak Dev University Amritsar Punjab 143005 India
- Drug and Pollution Testing Laboratory, Guru Nanak Dev University Amritsar Punjab 143005 India
| |
Collapse
|
16
|
Curieses Andrés CM, Pérez de la Lastra JM, Andrés Juan C, Plou FJ, Pérez-Lebeña E. From reactive species to disease development: Effect of oxidants and antioxidants on the cellular biomarkers. J Biochem Mol Toxicol 2023; 37:e23455. [PMID: 37437103 DOI: 10.1002/jbt.23455] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 06/14/2023] [Accepted: 06/29/2023] [Indexed: 07/14/2023]
Abstract
The influence of modern lifestyle, diet, exposure to chemicals such as phytosanitary substances, together with sedentary lifestyles and lack of exercise play an important role in inducing reactive stress (RS) and disease. The imbalance in the production and scavenging of free radicals and the induction of RS (oxidative, nitrosative, and halogenative) plays an essential role in the etiology of various chronic pathologies, such as cardiovascular diseases, diabetes, neurodegenerative diseases, and cancer. The implication of free radicals and reactive species injury in metabolic disturbances and the onset of many diseases have been accumulating for several decades, and are now accepted as a major cause of many chronic diseases. Exposure to elevated levels of free radicals can cause molecular structural impact on proteins, lipids, and DNA, as well as functional alteration of enzyme homeostasis, leading to aberrations in gene expression. Endogenous depletion of antioxidant enzymes can be mitigated using exogenous antioxidants. The current interest in the use of exogenous antioxidants as adjunctive agents for the treatment of human diseases allows a better understanding of these diseases, facilitating the development of new therapeutic agents with antioxidant activity to improve the treatment of various diseases. Here we examine the role that RS play in the initiation of disease and in the reactivity of free radicals and RS in organic and inorganic cellular components.
Collapse
Affiliation(s)
| | | | - Celia Andrés Juan
- Department of Organic Chemistry, Cinquima Institute, Faculty of Sciences, Valladolid University, Valladolid, Spain
| | - Francisco J Plou
- Institute of Catalysis and Petrochemistry, CSIC-Spanish Research Council, Madrid, Spain
| | | |
Collapse
|
17
|
DeVallance ER, Schmidt HM, Seman M, Lewis SE, Wood KC, Vickers SD, Hahn SA, Velayutham M, Hileman EA, Vitturi DA, Leonardi R, Straub AC, Kelley EE. Hemin and iron increase synthesis and trigger export of xanthine oxidoreductase from hepatocytes to the circulation. Redox Biol 2023; 67:102866. [PMID: 37703667 PMCID: PMC10506059 DOI: 10.1016/j.redox.2023.102866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/20/2023] [Accepted: 08/25/2023] [Indexed: 09/15/2023] Open
Abstract
We recently reported a previously unknown salutary role for xanthine oxidoreductase (XOR) in intravascular heme overload whereby hepatocellular export of XOR to the circulation was identified as a seminal step in affording protection. However, the cellular signaling and export mechanisms underpinning this process were not identified. Here, we present novel data showing hepatocytes upregulate XOR expression/protein abundance and actively release it to the extracellular compartment following exposure to hemopexin-bound hemin, hemin or free iron. For example, murine (AML-12 cells) hepatocytes treated with hemin (10 μM) exported XOR to the medium in the absence of cell death or loss of membrane integrity (2.0 ± 1.0 vs 16 ± 9 μU/mL p < 0.0001). The path of exocytosis was found to be noncanonical as pretreatment of the hepatocytes with Vaculin-1, a lysosomal trafficking inhibitor, and not Brefeldin A inhibited XOR release and promoted intracellular XOR accumulation (84 ± 17 vs 24 ± 8 hemin vs 5 ± 3 control μU/mg). Interestingly, free iron (Fe2+ and Fe3+) induced similar upregulation and release of XOR compared to hemin. Conversely, concomitant treatment with hemin and the classic transition metal chelator DTPA (20 μM) or uric acid completely blocked XOR release (p < 0.01). Our previously published time course showed XOR release from hepatocytes likely required transcriptional upregulation. As such, we determined that both Sp1 and NF-kB were acutely activated by hemin treatment (∼2-fold > controls for both, p < 0.05) and that silencing either or TLR4 with siRNA prevented hemin-induced XOR upregulation (p < 0.01). Finally, to confirm direct action of these transcription factors on the Xdh gene, chromatin immunoprecipitation was performed indicating that hemin significantly enriched (∼5-fold) both Sp1 and NF-kB near the transcription start site. In summary, our study identified a previously unknown pathway by which XOR is upregulated via SP1/NF-kB and subsequently exported to the extracellular environment. This is, to our knowledge, the very first study to demonstrate mechanistically that XOR can be specifically targeted for export as the seminal step in a compensatory response to heme/Fe overload.
Collapse
Affiliation(s)
- Evan R DeVallance
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA; Department of Physiology and Pharmacology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
| | - Heidi M Schmidt
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Madison Seman
- Department of Physiology and Pharmacology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
| | - Sara E Lewis
- Department of Physiology and Pharmacology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
| | - Katherine C Wood
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Schuyler D Vickers
- Department of Biochemistry, West Virginia University, Morgantown, WV, 26505, USA
| | - Scott A Hahn
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Murugesan Velayutham
- Department of Biochemistry, West Virginia University, Morgantown, WV, 26505, USA
| | - Emily A Hileman
- Department of Physiology and Pharmacology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
| | - Dario A Vitturi
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Roberta Leonardi
- Department of Biochemistry, West Virginia University, Morgantown, WV, 26505, USA
| | - Adam C Straub
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA; Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Eric E Kelley
- Department of Physiology and Pharmacology, Health Sciences Center, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
18
|
Matsuoka M, Yamaguchi J, Kinoshita K. Clinical Significance of Elevated Xanthine Dehydrogenase Levels and Hyperuricemia in Patients with Sepsis. Int J Mol Sci 2023; 24:13857. [PMID: 37762160 PMCID: PMC10530551 DOI: 10.3390/ijms241813857] [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: 07/03/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Patient outcomes for severe sepsis and septic shock remain poor. Excessive oxidative stress accelerates organ dysfunction in severe acute illnesses. Uric acid (UA) is the most abundant antioxidant. We hypothesized that UA and related molecules, which play a critical role in antioxidant activity, might be markers of oxidative stress in sepsis. The study aimed to clarify the clinical significance of UA and the relationship between UA, molecules related to UA, and outcomes by measuring blood UA, xanthine dehydrogenase (XDH), and 8-hydroxy-2-deoxyguanosine (8-OHdG) levels over time. Blood UA levels in septic patients were correlated with the SOFA score (ρ = 0.36, p < 0.0001) and blood XDH levels (ρ = 0.27, p < 0.0001). Blood XDH levels were correlated with the SOFA score (ρ = 0.59, p < 0.0001) and blood 8-OHdG levels (ρ = -0.32, p < 0.0001). Blood XDH levels were persistently high in fatal cases. Blood XDH level (OR 8.84, 95% CI: 1.42-91.2, p = 0.018) was an independent factor of poor outcomes. The cutoff of blood XDH level was 1.38 ng/mL (sensitivity 92.8%, specificity 61.9%), and those 1.38 ng/mL or higher were associated with a significantly reduced survival rate (blood XDH level > 1.38 ng/mL: 23.7%, blood XDH level < 1.38 ng/mL: 96.3%, respectively, p = 0.0007). Elevated UA levels due to elevated blood XDH levels in sepsis cases may reduce oxidative stress. Countermeasures against increased oxidative stress in sepsis may provide new therapeutic strategies.
Collapse
Affiliation(s)
| | - Junko Yamaguchi
- Division of Emergency and Critical Care Medicine, Department of Acute Medicine, Nihon University School of Medicine, 30-1, Oyaguchi Kami-cho, Itabashi-ku, Tokyo 173-8610, Japan; (M.M.); (K.K.)
| | | |
Collapse
|
19
|
Kohagura K, Satoh A, Kochi M, Nakamura T, Zamami R, Tana T, Kinjyo K, Funakoshi R, Yamazato M, Ishida A, Sakima A, Iseki K, Arima H, Ohya Y. Urate-lowering drugs for chronic kidney disease with asymptomatic hyperuricemia and hypertension: a randomized trial. J Hypertens 2023; 41:1420-1428. [PMID: 37334544 DOI: 10.1097/hjh.0000000000003484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
INTRODUCTION Xanthine oxidase (XO) inhibitors may slow down chronic kidney disease (CKD) progression. The comparative effectiveness of the different urate-lowering drugs is unknown. The aim of this study was to determine whether urate-lowering therapy with an XO inhibitor (febuxostat) and that with a uricosuric drug (benzbromarone) are comparable in slowing renal function decline in patients with CKD complicated with hypertension and hyperuricemia. METHODS This study was an open-label randomized parallel-group clinical trial of 95 patients with stage G3 CKD in Japan. The patients had hypertension and hyperuricemia without a history of gout. They were randomized to receive febuxostat ( n = 47; febuxostat group) or benzbromarone ( n = 48; benzbromarone group) and titrated to reduce their serum urate level to <6.0 mg/dl. The primary end-point was change in estimated glomerular filtration rate (eGFR) from baseline to 52 weeks. The secondary end-points included changes in uric acid level, blood pressure, urinary albumin-to-creatinine ratio, and XO activity. RESULTS Of the 95 patients, 88 (92.6%) completed the trial. There were no significant differences in change in eGFR (in ml/min/1.73 m 2 ) between the febuxostat [-0.23, 95% confidence interval (CI), -2.00 to 1.55] and benzbromarone (-2.18, 95% CI, -3.84 to -0.52) groups (difference, 1.95; 95% CI, -0.48 to 4.38; P = 0.115) nor in the secondary end-points, except for XO activity. Febuxostat significantly reduced XO activity ( P = 0.010). There were no significant differences in primary and secondary outcomes between the groups. A decrease in eGFR was significantly less in the febuxostat group than that of the benzbromarone group in the CKDG3a, but not in CKDG3b, in the subgroup analysis. There were no adverse effects specific to either drug. CONCLUSIONS No significant differences were found in the effects of febuxostat and benzbromarone in renal function decline in stage G3 CKD complicated with hyperuricemia and hypertension.
Collapse
Affiliation(s)
- Kentaro Kohagura
- Dialysis Unit, University of the Ryukyus Hospital, Nishihara-cho
| | - Atsushi Satoh
- Department of Preventive Medicine and Public Health, Fukuoka University, Fukuoka
| | - Masako Kochi
- Department of nephrology, Tomishiro Chuo Hospital, Tomigusuku
| | - Takuto Nakamura
- Department of Cardiovascular Medicine, Nephrology and Neurology, University of the Ryukyus Graduate School of Medicine, Nephrology and Neurology, Nishihara-cho
| | - Ryo Zamami
- Department of Cardiovascular Medicine, Nephrology and Neurology, University of the Ryukyus Graduate School of Medicine, Nephrology and Neurology, Nishihara-cho
| | | | | | - Ryo Funakoshi
- Department of internal medicine, Kaiho Hospital, Ginowan
| | - Masanobu Yamazato
- Department of Cardiovascular Medicine, Nephrology and Neurology, University of the Ryukyus Graduate School of Medicine, Nephrology and Neurology, Nishihara-cho
| | - Akio Ishida
- Department of Cardiovascular Medicine, Nephrology and Neurology, University of the Ryukyus Graduate School of Medicine, Nephrology and Neurology, Nishihara-cho
| | - Atsushi Sakima
- Health Administration Center, University of the Ryukyus, Nishihara-cho
| | | | - Hisatomi Arima
- Department of Preventive Medicine and Public Health, Fukuoka University, Fukuoka
| | - Yusuke Ohya
- Department of Cardiovascular Medicine, Nephrology and Neurology, University of the Ryukyus Graduate School of Medicine, Nephrology and Neurology, Nishihara-cho
| |
Collapse
|
20
|
Zhang J, Han L, Liu H, Zhang H, An Z. Metabolomic analysis reveals the metabolic disturbance in aortic dissection: Subtype difference and accurate diagnosis. Nutr Metab Cardiovasc Dis 2023; 33:1556-1564. [PMID: 37263915 DOI: 10.1016/j.numecd.2023.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 04/12/2023] [Accepted: 05/03/2023] [Indexed: 06/03/2023]
Abstract
BACKGROUND AND AIMS Aortic dissection (AD), a severe clinical emergency with high mortality, is easily misdiagnosed as are other cardiovascular diseases. This study aimed at discovering plasma metabolic markers with the potential to diagnose AD and clarifying the metabolic differences between two subtypes of AD. METHODS AND RESULTS To facilitate the diagnosis of AD, we investigated the plasma metabolic profile by metabolomic approach. A total 482 human subjects were enrolled in the study: 80 patients with AD (50 with Stanford type A and 30 with Stanford type B), 198 coronary artery disease (CAD) patients, and 204 healthy individuals. Plasma samples were submitted to targeted metabolomic analysis. The partial least-squares discriminant analysis models were constructed to illustrate clear discrimination of AD patients with CAD patients and healthy control. Subsequently, the metabolites that were clinically relevant to the disturbances in AD were identified. Twenty metabolites induced the separation of AD patients and healthy control, 9 of which caused the separation of CAD patients and healthy control. There are 11 metabolites specifically down-regulated in AD group. Subgroup analysis showed that the levels of glycerol and uridine were dramatically lower in the plasma of patients with Stanford type A AD than those in the healthy control or Stanford type B AD groups. CONCLUSION This study characterized metabolomic profiles specifically associated with the pathogenesis and development of AD. The findings of this research may potentially lead to earlier diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Jinghui Zhang
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100029, China
| | - Lu Han
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100029, China; Beijing Lab for Cardiovascular Precision Medicine, Beijing, 100069, China; Key Laboratory of Medical Engineering for Cardiovascular Disease, Beijing, 100069, China
| | - Hongchuan Liu
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100029, China
| | - Hongjia Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China; Beijing Lab for Cardiovascular Precision Medicine, Beijing, 100069, China; Key Laboratory of Medical Engineering for Cardiovascular Disease, Beijing, 100069, China.
| | - Zhuoling An
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100029, China.
| |
Collapse
|
21
|
Li Y, Feng Y, Yang Z, Zhou Z, Jiang D, Luo J. Untargeted metabolomics of saliva in pregnant women with and without gestational diabetes mellitus and healthy non-pregnant women. Front Cell Infect Microbiol 2023; 13:1206462. [PMID: 37538307 PMCID: PMC10394705 DOI: 10.3389/fcimb.2023.1206462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 06/23/2023] [Indexed: 08/05/2023] Open
Abstract
Objective The aim of this study was to compare the differences in salivary metabolites between pregnant women with gestational diabetes mellitus (GDM), healthy pregnant women (HPW), and healthy non-pregnant women (HNPW), and analyze the possible associations between the identified metabolites and gingivitis. Method The study included women with GDM (n = 9, mean age 28.9 ± 3.6 years, mean gestational age 30.1 ± 3.2 weeks), HPW (n = 9, mean age 27.9 ± 3.0 years, mean gestational age 28.6 ± 4.7 weeks), and HNPW (n = 9, mean age 27.7 ± 2.1 years). Saliva samples were collected from all participants and were analyzed with LC-MS/MS-based untargeted metabolomic analysis. Metabolite extraction, qualitative and semi-quantitative analysis, and bioinformatics analysis were performed to identify the differential metabolites and metabolic pathways between groups. The identified differential metabolites were further analyzed in an attempt to explore their possible associations with periodontal health and provide evidence for the prevention and treatment of periodontal inflammation during pregnancy. Results In positive ion mode, a total of 2,529 molecular features were detected in all samples, 166 differential metabolites were identified between the GDM and HPW groups (89 upregulated and 77 downregulated), 823 differential metabolites were identified between the GDM and HNPW groups (402 upregulated and 421 downregulated), and 647 differential metabolites were identified between the HPW and HNPW groups (351 upregulated and 296 downregulated). In negative ion mode, 983 metabolites were detected in all samples, 49 differential metabolites were identified between the GDM and HPW groups (29 upregulated and 20 downregulated), 341 differential metabolites were identified between the GDM and HNPW groups (167 upregulated and 174 downregulated), and 245 differential metabolites were identified between the HPW and HNPW groups (112 upregulated and 133 downregulated). A total of nine differential metabolites with high confidence levels were identified in both the positive and negative ion modes, namely, L-isoleucine, D-glucose 6-phosphate, docosahexaenoic acid, arachidonic acid, adenosine, adenosine-monophosphate, adenosine 5'-monophosphate, xanthine, and hypoxanthine. Among all pathways enriched by the upregulated differential metabolites, the largest number of pathways were enriched by four differential metabolites, adenosine, adenosine 5'-monophosphate, D-glucose 6-phosphate, and adenosine-monophosphate, and among all pathways enriched by the downregulated differential metabolites, the largest number of pathways were enriched by three differential metabolites, L-isoleucine, xanthine, and arachidonic acid. Conclusion Untargeted metabolomic analysis of saliva samples from pregnant women with GDM, HPW, and HNPW identified nine differential metabolites with high confidence. The results are similar to findings from previous metabolomics studies of serum and urine samples, which offer the possibility of using saliva for regular noninvasive testing in the population of pregnant women with and without GDM. Meanwhile, the associations between these identified differential metabolites and gingivitis need to be further validated by subsequent studies.
Collapse
Affiliation(s)
- Yueheng Li
- Department of Preventive Dentistry, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yang Feng
- Chongqing Changshou Health Center for Women and Children, Chongqing, China
| | - Zhengyan Yang
- Department of Preventive Dentistry, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Zhi Zhou
- Department of Preventive Dentistry, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Dan Jiang
- Department of Preventive Dentistry, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jun Luo
- Department of Preventive Dentistry, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
22
|
Gao L, Rafaels N, Dudenkov TM, Damarla M, Damico R, Maloney JP, Moss M, Martin GS, Sevransky J, Shanholtz C, Herr DL, Garcia JGN, Hernandez-Beeftink T, Villar J, Flores C, Beaty TH, Brower R, Hassoun PM, Barnes KC. Xanthine oxidoreductase gene polymorphisms are associated with high risk of sepsis and organ failure. Respir Res 2023; 24:177. [PMID: 37415141 PMCID: PMC10324226 DOI: 10.1186/s12931-023-02481-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/20/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND Sepsis and associated organ failures confer substantial morbidity and mortality. Xanthine oxidoreductase (XOR) is implicated in the development of tissue oxidative damage in a wide variety of respiratory and cardiovascular disorders including sepsis and sepsis-associated acute respiratory distress syndrome (ARDS). We examined whether single nucleotide polymorphisms (SNPs) in the XDH gene (encoding XOR) might influence susceptibility to and outcome in patients with sepsis. METHODS We genotyped 28 tag SNPs in XDH gene in the CELEG cohort, including 621 European American (EA) and 353 African American (AA) sepsis patients. Serum XOR activity was measured in a subset of CELEG subjects. Additionally, we assessed the functional effects of XDH variants utilizing empirical data from different integrated software tools and datasets. RESULTS Among AA patients, six intronic variants (rs206805, rs513311, rs185925, rs561525, rs2163059, rs13387204), in a region enriched with regulatory elements, were associated with risk of sepsis (P < 0.008-0.049). Two out of six SNPs (rs561525 and rs2163059) were associated with risk of sepsis-associated ARDS in an independent validation cohort (GEN-SEP) of 590 sepsis patients of European descent. Two common SNPs (rs1884725 and rs4952085) in tight linkage disequilibrium (LD) provided strong evidence for association with increased levels of serum creatinine (Padjusted<0.0005 and 0.0006, respectively), suggesting a role in increased risk of renal dysfunction. In contrast, among EA ARDS patients, the missense variant rs17011368 (I703V) was associated with enhanced mortality at 60-days (P < 0.038). We found higher serum XOR activity in 143 sepsis patients (54.5 ± 57.1 mU/mL) compared to 31 controls (20.9 ± 12.4 mU/mL, P = 1.96 × 10- 13). XOR activity was associated with the lead variant rs185925 among AA sepsis patients with ARDS (P < 0.005 and Padjusted<0.01). Multifaceted functions of prioritized XDH variants, as suggested by various functional annotation tools, support their potential causality in sepsis. CONCLUSIONS Our findings suggest that XOR is a novel combined genetic and biochemical marker for risk and outcome in patients with sepsis and ARDS.
Collapse
Affiliation(s)
- Li Gao
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- The Johns Hopkins Asthma & Allergy Center, 5501 Hopkins Bayview Circle, Room 3B.65B, Baltimore, MD, 21224, USA.
| | - Nicholas Rafaels
- Division of Biomedical Informatics & Personalized Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Tanda M Dudenkov
- Department of Epidemiology, Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, MD, USA
| | - Mahendra Damarla
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rachel Damico
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James P Maloney
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Marc Moss
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Greg S Martin
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Jonathan Sevransky
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Carl Shanholtz
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dan L Herr
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joe G N Garcia
- University of Arizona College of Medicine, Tucson, AZ, USA
| | - Tamara Hernandez-Beeftink
- Research Unit, Hospital Universitario Ntra. Sra. de Candelaria, Santa Cruz de Tenerife, Spain
- Research Unit, Hospital Universitario Dr. Negrin, Las Palmas de Gran Canaria, Spain
| | - Jesús Villar
- Research Unit, Hospital Universitario Dr. Negrin, Las Palmas de Gran Canaria, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Li Ka Shing Knowledge Institiute at St. Michael's Hospital, Toronto, Canada
| | - Carlos Flores
- Research Unit, Hospital Universitario Ntra. Sra. de Candelaria, Santa Cruz de Tenerife, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
- Facultad de Ciencias de la Salud, Universidad Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - Terri H Beaty
- Department of Epidemiology, Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, MD, USA
| | - Roy Brower
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul M Hassoun
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- The Johns Hopkins Asthma & Allergy Center, 5501 Hopkins Bayview Circle, Room 3B.65B, Baltimore, MD, 21224, USA.
| | - Kathleen C Barnes
- Division of Biomedical Informatics & Personalized Medicine, University of Colorado School of Medicine, Aurora, CO, USA.
- University of Colorado Anschutz Medical Campus, 13001 E. 17th Place, Room 5330A, Aurora, CO, 80045, USA.
| |
Collapse
|
23
|
Al-Dulaimy WYM, Hussein AA, Mahdi MA, Kadhom M. In Vitro Inhibition of Xanthine Oxidase Purified from Arthritis Serum Patients by Nanocurcumin and Artemisinin Active Compounds. Molecules 2023; 28:5124. [PMID: 37446786 DOI: 10.3390/molecules28135124] [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: 05/15/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Curcumin and artemisinin are commonly used in traditional East Asian medicine. In this study, we investigated the inhibitory effects of these active compounds on xanthine oxidase (XO) using allopurinol as a control. XO was purified from the serum of arthritis patients through ammonium sulfate precipitation (65%) and ion exchange chromatography on diethylaminoethyl (DEAE)-cellulose. The specific activity of the purified enzyme was 32.5 U/mg protein, resulting in a 7-fold purification with a yield of 66.8%. Molecular docking analysis revealed that curcumin had the strongest interaction energy with XO, with a binding energy of -9.28 kcal/mol. The amino acid residues Thr1077, Gln762, Phe914, Ala1078, Val1011, Glu1194, and Ala1079 were located closer to the binding site of curcumin than artemisinin, which had a binding energy of -7.2 kcal/mol. In vitro inhibition assays were performed using nanocurcumin and artemisinin at concentrations of 5, 10, 15, 20, and 25 µg/mL. Curcumin inhibited enzyme activity by 67-91%, while artemisinin had a lower inhibition ratio, which ranged from 40-70% compared to allopurinol as a control.
Collapse
Affiliation(s)
| | - Asmaa A Hussein
- Department of Molecular and Medical Biotechnology, College of Biotechnology, Al-Nahrain University, Jadriya, Baghdad 64074, Iraq
| | - Mohammed Asaad Mahdi
- Department of Chemistry, College of Science, University of Diyala, Baquba 32001, Iraq
| | - Mohammed Kadhom
- Department of Environmental Science, College of Energy and Environmental Science, Al-Karkh University of Science, Baghdad 10081, Iraq
| |
Collapse
|
24
|
Zhang WZ. Uric acid en route to gout. Adv Clin Chem 2023; 116:209-275. [PMID: 37852720 DOI: 10.1016/bs.acc.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Gout and hyperuricemia (HU) have generated immense attention due to increased prevalence. Gout is a multifactorial metabolic and inflammatory disease that occurs when increased uric acid (UA) induce HU resulting in monosodium urate (MSU) crystal deposition in joints. However, gout pathogenesis does not always involve these events and HU does not always cause a gout flare. Treatment with UA-lowering therapeutics may not prevent or reduce the incidence of gout flare or gout-associated comorbidities. UA exhibits both pro- and anti-inflammation functions in gout pathogenesis. HU and gout share mechanistic and metabolic connections at a systematic level, as shown by studies on associated comorbidities. Recent studies on the interplay between UA, HU, MSU and gout as well as the development of HU and gout in association with metabolic syndromes, non-alcoholic fatty liver disease (NAFLD), and cardiovascular, renal and cerebrovascular diseases are discussed. This review examines current and potential therapeutic regimens and illuminates the journey from disrupted UA to gout.
Collapse
Affiliation(s)
- Wei-Zheng Zhang
- VIDRL, The Peter Doherty Institute, Melbourne, VIC, Australia.
| |
Collapse
|
25
|
Liu Y, Croft KD, Mori TA, Gaspari TA, Kemp-Harper BK, Ward NC. Long-term dietary nitrate supplementation slows the progression of established atherosclerosis in ApoE -/- mice fed a high fat diet. Eur J Nutr 2023; 62:1845-1857. [PMID: 36853380 PMCID: PMC10195750 DOI: 10.1007/s00394-023-03127-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/20/2023] [Indexed: 03/01/2023]
Abstract
BACKGROUND AND AIMS Atherosclerosis is associated with a reduction in the bioavailability and/or bioactivity of endogenous nitric oxide (NO). Dietary nitrate has been proposed as an alternate source when endogenous NO production is reduced. Our previous study demonstrated a protective effect of dietary nitrate on the development of atherosclerosis in the apoE-/- mouse model. However most patients do not present clinically until well after the disease is established. The aims of this study were to determine whether chronic dietary nitrate supplementation can prevent or reverse the progression of atherosclerosis after disease is already established, as well as to explore the underlying mechanism of these cardiovascular protective effects. METHODS 60 apoE-/- mice were given a high fat diet (HFD) for 12 weeks to allow for the development of atherosclerosis. The mice were then randomized to (i) control group (HFD + 1 mmol/kg/day NaCl), (ii) moderate-dose group (HFD +1 mmol/kg/day NaNO3), or (iii) high-dose group (HFD + 10 mmol/kg/day NaNO3) (20/group) for a further 12 weeks. A group of apoE-/- mice (n = 20) consumed a normal laboratory chow diet for 24 weeks and were included as a reference group. RESULTS Long-term supplementation with high dose nitrate resulted in ~ 50% reduction in plaque lesion area. Collagen expression and smooth muscle accumulation were increased, and lipid deposition and macrophage accumulation were reduced within atherosclerotic plaques of mice supplemented with high dose nitrate. These changes were associated with an increase in nitrite reductase as well as activation of the endogenous eNOS-NO pathway. CONCLUSION Long-term high dose nitrate significantly attenuated the progression of established atherosclerosis in the apoE-/- mice fed a HFD. This appears to be mediated in part through a XOR-dependent reduction of nitrate to NO, as well as enhanced eNOS activation via increased Akt and eNOS phosphorylation.
Collapse
Affiliation(s)
- Yang Liu
- School of Biomedical Sciences, University of Western Australia, Perth, WA Australia
| | - Kevin D. Croft
- School of Biomedical Sciences, University of Western Australia, Perth, WA Australia
| | - Trevor A. Mori
- Medical School, University of Western Australia, Perth, WA Australia
| | - Tracey A. Gaspari
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC Australia
| | - Barbara K. Kemp-Harper
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC Australia
| | - Natalie C. Ward
- Medical School, University of Western Australia, Perth, WA Australia
- Dobney Hypertension Centre, Medical School, University of Western Australia, G.P.O Box X2213, Perth, WA 6847 Australia
| |
Collapse
|
26
|
Yu H, Song YY, Li XH. Early diabetic kidney disease: Focus on the glycocalyx. World J Diabetes 2023; 14:460-480. [PMID: 37273258 PMCID: PMC10236994 DOI: 10.4239/wjd.v14.i5.460] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/10/2023] [Accepted: 04/12/2023] [Indexed: 05/15/2023] Open
Abstract
The incidence of diabetic kidney disease (DKD) is sharply increasing worldwide. Microalbuminuria is the primary clinical marker used to identify DKD, and its initiating step in diabetes is glomerular endothelial cell dysfunction, particularly glycocalyx impairment. The glycocalyx found on the surface of glomerular endothelial cells, is a dynamic hydrated layer structure composed of pro-teoglycans, glycoproteins, and some adsorbed soluble components. It reinforces the negative charge barrier, transduces the shear stress, and mediates the interaction of blood corpuscles and podocytes with endothelial cells. In the high-glucose environment of diabetes, excessive reactive oxygen species and proinflammatory cytokines can damage the endothelial glycocalyx (EG) both directly and indirectly, which induces the production of microalbuminuria. Further research is required to elucidate the role of the podocyte glycocalyx, which may, together with endothelial cells, form a line of defense against albumin filtration. Interestingly, recent research has confirmed that the negative charge barrier function of the glycocalyx found in the glomerular basement membrane and its repulsion effect on albumin is limited. Therefore, to improve the early diagnosis and treatment of DKD, the potential mechanisms of EG degradation must be analyzed and more responsive and controllable targets must be explored. The content of this review will provide insights for future research.
Collapse
Affiliation(s)
- Hui Yu
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Yi-Yun Song
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Xian-Hua Li
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| |
Collapse
|
27
|
Tian X, Wang P, Chen S, Zhang Y, Zhang X, Xu Q, Luo Y, Wu S, Wang A. Association of Normal Serum Uric Acid Level and Cardiovascular Disease in People Without Risk Factors for Cardiac Diseases in China. J Am Heart Assoc 2023; 12:e029633. [PMID: 37183869 DOI: 10.1161/jaha.123.029633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Background Healthy individuals with normal level of serum uric acid (SUA) may not be truly at the lowest risk of cardiovascular disease (CVD). This study aimed to assess the association of SUA levels with CVD and its subtypes in people without CVD risk factors and determine a suitable target of SUA to prevent CVD. Methods and Results We enrolled 25 284 participants who were free of CVD, absent of CVD risk factors, and with an SUA level between 180 and 359 μmol/L (3-6 mg/dL) at baseline from the Kailuan study. Cox proportional hazards models were applied to calculated adjusted hazard ratio (HR) and 95% CI for the risk of CVD and its subtypes. During a median follow-up of 12.97 years (interquartile range, 12.68-13.16 years), we identified 1007 cases of CVD. There was an increase in the risk of incident CVD with increasing SUA levels (Ptrend=0.0011). Compared with participants with SUA levels of 180 to 239 μmol/L (3-4 mg/dL), the HR of CVD was 1.12 (95% CI, 0.96-1.31) and 1.28 (95% CI, 1.08-1.52) for SUA levels of 240 to 299 μmol/L (4-5 mg/dL) and 300 to 359 μmol/L (5-6 mg/dL), respectively. A multivariable-adjusted spline regression model showed a J-shaped association between SUA and the risk of CVD. Similar results were observed for stroke and myocardial infarction. Conclusions The risk of incident CVD increased with elevating SUA levels among individuals without hyperuricemia or other traditional CVD risk factors. These findings highlighted the importance of primordial prevention for SUA level increase along with other traditional CVD risk factors.
Collapse
Affiliation(s)
- Xue Tian
- Department of Neurology, Beijing Tiantan Hospital Capital Medical University Beijing China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital Capital Medical University Beijing China
- Department of Epidemiology and Health Statistics, School of Public Health Capital Medical University Beijing China
- Beijing Municipal Key Laboratory of Clinical Epidemiology Beijing China
| | - Penglian Wang
- Department of Neurology, Beijing Tiantan Hospital Capital Medical University Beijing China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital Capital Medical University Beijing China
| | - Shuohua Chen
- Department of Cardiology, Kailuan Hospital North China University of Science and Technology Tangshan China
| | - Yijun Zhang
- Department of Neurology, Beijing Tiantan Hospital Capital Medical University Beijing China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital Capital Medical University Beijing China
- Department of Epidemiology and Health Statistics, School of Public Health Capital Medical University Beijing China
- Beijing Municipal Key Laboratory of Clinical Epidemiology Beijing China
| | - Xiaoli Zhang
- Department of Neurology, Beijing Tiantan Hospital Capital Medical University Beijing China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital Capital Medical University Beijing China
| | - Qin Xu
- Department of Neurology, Beijing Tiantan Hospital Capital Medical University Beijing China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital Capital Medical University Beijing China
| | - Yanxia Luo
- Department of Epidemiology and Health Statistics, School of Public Health Capital Medical University Beijing China
- Beijing Municipal Key Laboratory of Clinical Epidemiology Beijing China
| | - Shouling Wu
- Department of Cardiology, Kailuan Hospital North China University of Science and Technology Tangshan China
| | - Anxin Wang
- Department of Neurology, Beijing Tiantan Hospital Capital Medical University Beijing China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital Capital Medical University Beijing China
| |
Collapse
|
28
|
Sato K, Naganuma A, Nagashima T, Arai Y, Mikami Y, Nakajima Y, Kanayama Y, Murakami T, Uehara S, Uehara D, Yamazaki Y, Murase T, Nakamura T, Uraoka T. A Newly Developed Method-Based Xanthine Oxidoreductase Activities in Various Human Liver Diseases. Biomedicines 2023; 11:biomedicines11051445. [PMID: 37239117 PMCID: PMC10216503 DOI: 10.3390/biomedicines11051445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/03/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Studies evaluating xanthine oxidoreductase (XOR) activities in comprehensive liver diseases are scarce, and different etiologies have previously been combined in groups for comparison. To accurately evaluate XOR activities in liver diseases, the plasma XOR activities in etiology-based comprehensive liver diseases were measured using a novel, sensitive, and accurate assay that is a combination of liquid chromatography and triple quadrupole mass spectrometry to detect [13C2, 15N2]uric acid using [13C2, 15N2]xanthine as a substrate. We also mainly evaluated the association between the plasma XOR activities and parameters of liver tests, purine metabolism-associated markers, oxidative stress markers, and an inflammation marker. In total, 329 patients and 32 controls were enrolled in our study. Plasma XOR activities were generally increased in liver diseases, especially in the active phase, such as in patients with hepatitis C virus RNA positivity, those with abnormal alanine transaminase (ALT) levels in autoimmune liver diseases, and uncured hepatocellular carcinoma patients. Plasma XOR activities were numerically highest in patients with acute hepatitis B. Plasma XOR activities were closely correlated with parameters of liver tests, especially serum ALT levels, regardless of etiology and plasma xanthine levels. Our results indicated that plasma XOR activity might reflect the active phase in various liver diseases.
Collapse
Affiliation(s)
- Ken Sato
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
- Department of Hepatology, Heisei Hidaka Clinic, Takasaki 371-0001, Japan
- Department of Healthcare Informatics, Takasaki University of Health and Welfare, Takasaki 370-0033, Japan
| | - Atsushi Naganuma
- Department of Gastroenterology, National Hospital Organization Takasaki General Medical Center, Takasaki 370-0829, Japan
| | - Tamon Nagashima
- Department of Gastroenterology, National Hospital Organization Shibukawa Medical Center, Shibukawa 377-0204, Japan
| | - Yosuke Arai
- Department of Gastroenterology, National Hospital Organization Shibukawa Medical Center, Shibukawa 377-0204, Japan
| | - Yuka Mikami
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Yuka Nakajima
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Yuki Kanayama
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Tatsuma Murakami
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
- Department of Gastroenterology, National Hospital Organization Takasaki General Medical Center, Takasaki 370-0829, Japan
| | - Sanae Uehara
- Department of Gastroenterology, National Hospital Organization Takasaki General Medical Center, Takasaki 370-0829, Japan
| | - Daisuke Uehara
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Yuichi Yamazaki
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Takayo Murase
- Mie Research Park, Sanwa Kagaku Kenkyusho, Inabe 511-0406, Japan
| | - Takashi Nakamura
- Mie Research Park, Sanwa Kagaku Kenkyusho, Inabe 511-0406, Japan
| | - Toshio Uraoka
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| |
Collapse
|
29
|
Kotozaki Y, Satoh M, Nasu T, Tanno K, Tanaka F, Sasaki M. Human Plasma Xanthine Oxidoreductase Activity in Cardiovascular Disease: Evidence from a Population-Based Study. Biomedicines 2023; 11:biomedicines11030754. [PMID: 36979733 PMCID: PMC10045414 DOI: 10.3390/biomedicines11030754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/20/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Xanthine oxidoreductase (XOR) and its products contribute to the development of chronic inflammation and oxidative stress. Excessive XOR activity is believed to promote inflammatory responses and atherosclerotic plaque formation, which are major cardiovascular risk factors. The mechanisms of XOR activity in the development and progression of cardiovascular disease (CVD), coupled with the complexity of the relationship between XOR activity and the biological effects of uric acid; reactive oxygen species; and nitric oxide, which are the major products of XOR activity, have long been debated, but have not yet been clearly elucidated. Recently, a system for measuring highly sensitive XOR activity in human plasma was established, and there has been progress in the research on the mechanisms of XOR activity. In addition, there are accumulating findings about the relationship between XOR activity and CVD. In this narrative review, we summarize existing knowledge regarding plasma XOR activity and its relationship with CVD and discuss future perspectives.
Collapse
Affiliation(s)
- Yuka Kotozaki
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, 1-1-1, Idaidori, Yahaba 028-3694, Iwate, Japan
- Correspondence: (Y.K.); (M.S.)
| | - Mamoru Satoh
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, 1-1-1, Idaidori, Yahaba 028-3694, Iwate, Japan
- Department of Biomedical Information Analysis, Institute for Biomedical Sciences, Iwate Medical University, 1-1-1, Idaidori, Yahaba 028-3694, Iwate, Japan
- Correspondence: (Y.K.); (M.S.)
| | - Takahito Nasu
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, 1-1-1, Idaidori, Yahaba 028-3694, Iwate, Japan
- Department of Biomedical Information Analysis, Institute for Biomedical Sciences, Iwate Medical University, 1-1-1, Idaidori, Yahaba 028-3694, Iwate, Japan
- Division of Cardiology, Department of Internal Medicine, Iwate Medical University, 2-1-1, Idaidori, Yahaba 028-3694, Iwate, Japan
| | - Kozo Tanno
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, 1-1-1, Idaidori, Yahaba 028-3694, Iwate, Japan
- Department of Hygiene and Preventive Medicine, Iwate Medical University, 1-1-1, Idaidori, Yahaba 028-3694, Iwate, Japan
| | - Fumitaka Tanaka
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, 1-1-1, Idaidori, Yahaba 028-3694, Iwate, Japan
- Division of Nephrology and Hypertension, Department of Internal Medicine, Iwate Medical University, 2-1-1, Idaidori, Yahaba 028-3694, Iwate, Japan
| | - Makoto Sasaki
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, 1-1-1, Idaidori, Yahaba 028-3694, Iwate, Japan
- Division of Ultrahigh field MRI, Institute for Biomedical Sciences, Iwate Medical University, 1-1-1, Idaidori, Yahaba 028-3694, Iwate, Japan
| |
Collapse
|
30
|
The Effects of Topiroxostat, a Selective Xanthine Oxidoreductase Inhibitor, on Arterial Stiffness in Hyperuricemic Patients with Liver Dysfunction: A Sub-Analysis of the BEYOND-UA Study. Biomedicines 2023; 11:biomedicines11030674. [PMID: 36979653 PMCID: PMC10045538 DOI: 10.3390/biomedicines11030674] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Background: The effects of uric acid (UA)-lowering therapy with xanthine oxidoreductase (XOR) inhibitors on the development of cardiovascular diseases remain controversial. Based on recent findings that plasma XOR activity increased in liver disease conditions, we conducted a sub-analysis of the BEYOND-UA study to examine the differential effects of topiroxostat on arterial stiffness based on liver function in hyperuricemic individuals with hypertension. Methods: Sixty-three subjects treated with topiroxostat were grouped according to baseline alanine aminotransferase (ALT) levels (above or below cut-off values of 22, 30, or 40 U/L). The primary endpoint was changes in the cardio-ankle vascular index (CAVI) from baseline to 24 weeks. Results: Significant reductions in CAVI during topiroxostat therapy occurred in subjects with baseline ALT ≥30 U/L or ≥40 U/L, and significant between-group differences were detected. Brachial-ankle pulse wave velocity significantly decreased in the ALT-high groups at all cut-off values. Reductions in morning home blood pressure and serum UA were similar regardless of the baseline ALT level. For eleven subjects with available data, ALT-high groups showed high plasma XOR activity, which was significantly suppressed by topiroxostat. Conclusions: Topiroxostat improved arterial stiffness parameters in hyperuricemic patients with liver dysfunction, which might be related to its inhibitory effect on plasma XOR.
Collapse
|
31
|
Watanabe K, Arimoto T, Watanabe T, Otaki Y, Murase T, Nakamura T, Kobayashi Y, Aono T, Saito Y, Koyama K, Hashimoto N, Kutsuzawa D, Kato S, Tamura H, Nishiyama S, Takahashi H, Watanabe M. Prognostic impact of plasma xanthine oxidoreductase activity in patients with heart failure with atrial fibrillation. J Cardiol 2023; 81:469-475. [PMID: 36822544 DOI: 10.1016/j.jjcc.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUND Xanthine oxidoreductase (XOR) is a rate-limiting enzyme for uric acid (UA) production and plays an important role in generating reactive oxygen species (ROS). Overproduction of ROS is reported to contribute to the pathophysiology of atrial fibrillation (AF), however, the prognostic impact of plasma XOR activity in patients with heart failure (HF) with AF is undetermined. METHODS We measured plasma XOR activity in 475 HF patients, including those with sinus rhythm (HF-SR, n = 211), and those with AF (HF-AF, n = 264). The type of AF included paroxysmal (n = 128) and persistent (n = 136) AF. All patients were prospectively followed up for a median period of 804 days. RESULTS HF-AF patients had significantly higher plasma XOR activity and serum UA levels compared with HF-SR patients. Both plasma XOR activity and serum UA levels were higher in patients with persistent AF than in those with SR and with paroxysmal AF. Multivariate linear regression analysis showed that persistent AF was independently associated with increased XOR activity. During the follow-up period, there were 79 major adverse cardiovascular events (MACEs). HF-AF patients with MACEs had higher plasma XOR activity compared with those without MACEs, while there were no significant differences in serum UA levels. Multivariate Cox proportional analysis showed that high XOR activity was an independent risk factor for MACEs after adjustment for confounding factors. Kaplan-Meier analysis revealed that the high XOR activity group had a higher risk of MACEs than the low XOR activity group. The prediction model was significantly improved by the addition of XOR activity to the basic predictors. CONCLUSIONS HF-AF patients had significantly higher plasma XOR activity compared with HF-SR patients. Plasma XOR activity proved to be a reliable indicator for MACEs in HF-AF patients.
Collapse
Affiliation(s)
- Ken Watanabe
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan
| | - Takanori Arimoto
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan.
| | - Tetsu Watanabe
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan
| | - Yoichiro Otaki
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan
| | - Takayo Murase
- Mie Research Laboratories, Sanwa Kagaku Kenkyusho Co., Ltd., Mie, Japan
| | - Takashi Nakamura
- Mie Research Laboratories, Sanwa Kagaku Kenkyusho Co., Ltd., Mie, Japan
| | - Yuta Kobayashi
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan
| | - Tomonori Aono
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan
| | - Yuji Saito
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan
| | - Kyoko Koyama
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan
| | - Naoaki Hashimoto
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan
| | - Daisuke Kutsuzawa
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan
| | - Shigehiko Kato
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan
| | - Harutoshi Tamura
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan
| | - Satoshi Nishiyama
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan
| | - Hiroki Takahashi
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan
| | - Masafumi Watanabe
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, Yamagata, Japan
| |
Collapse
|
32
|
Noda M, Kikuchi C, Tarui R, Nakamura T, Murase T, Hori E, Matsunaga T. Effect of Topiroxostat on Reducing Oxidative Stress in the Aorta of Streptozotocin-Induced Diabetic Rats. Biol Pharm Bull 2023; 46:272-278. [PMID: 36529499 DOI: 10.1248/bpb.b22-00694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Xanthine oxidoreductase exists both intracellularly and extracellularly and induces vascular injury by producing reactive oxygen species (ROS). Here, we investigated the effects and mechanism of action of topiroxostat, a xanthine oxidase inhibitor, on ROS using an animal model of type 1 diabetes with persistent hyperglycemia. Six-week-old male Sprague-Dawley rats were administered 50 mg/kg streptozotocin to induce diabetes; at 8 weeks of age, animals were administered topiroxostat (0.3, 1, or 3 mg/kg) for 2 weeks through mixed feeding after which the aorta was sampled. The production of superoxide, a type of ROS, was measured by chemiluminescence and dihydroethidium staining. Cytotoxicity was evaluated by nitrotyrosine staining. Topiroxostat at 3 mg/kg significantly decreased blood urea nitrogen, e-selectin, urinary malondialdehyde, and the urinary albumin/creatinine ratio compared with the streptozotocin group. Superoxide production by xanthine oxidase anchored to the cell membrane was significantly decreased by topiroxostat at both 1 mg/kg and 3 mg/kg compared with the streptozotocin group. Dihydroethidium staining revealed no significant effect of topiroxostat administration on superoxide production. The fluorescence intensity of nitrotyrosine staining was significantly suppressed by 3 mg/kg topiroxostat. Topiroxostat was found to inhibit the production of ROS in the thoracic aorta and suppress vascular endothelial damage. The antioxidant effect of topiroxostat appears to be exerted via the inhibition of anchored xanthine oxidase.
Collapse
Affiliation(s)
- Masato Noda
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Chigusa Kikuchi
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University.,Laboratory of Community Medicine, Showa Pharmaceutical University.,Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University
| | - Ryota Tarui
- Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University
| | - Takashi Nakamura
- Pharmacological Study Group, Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho
| | - Takayo Murase
- Pharmacological Study Group, Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho
| | - Eisei Hori
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University.,Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University
| | - Tamihide Matsunaga
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University.,Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University
| |
Collapse
|
33
|
Implication of changes in xanthine oxidase activity following hemodialysis. BMC Nephrol 2023; 24:13. [PMID: 36647052 PMCID: PMC9841642 DOI: 10.1186/s12882-023-03062-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/29/2022] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Xanthine oxidase activity has a key role in the development of oxidative stress and progression of cardiovascular diseases. However, the change of xanthine oxidase activity following hemodialysis and its prognostic impact remain uncertain. METHODS We prospectively included hemodialysis patients who did not take any anti-hyperuricemic agents and measured their xanthine oxidase activity before and after the index hemodialysis. The impact of change in xanthine oxidase activity during hemodialysis on cardiovascular death were investigated. RESULTS A total of 46 patients (median 72 years old, 29 men) were included. During hemodialysis, a common logarithm of xanthine oxidase activity decreased significantly from 1.16 (0.94, 1.27) to 1.03 (0.80, 1.20) (p < 0.01). Of them, xanthine oxidase activity remained unchanged or increased in 16 patients, who had a greater decrease in blood pressure and more hemoconcentration compared with others. Two-year survival from cardiovascular death was not significantly stratified by the changes in xanthine oxidase activity (p = 0.43). CONCLUSIONS During hemodialysis, xanthine oxidase activity decreased among the overall cohort, whereas some patients experienced its increases, which might be associated with hypotension and hemoconcentration during hemodialysis. Further larger-scale studies are required to validate our findings and find clinical implication of change in xanthine oxidase activity during hemodialysis.
Collapse
|
34
|
Lind L, Fall T, Ärnlöv J, Elmståhl S, Sundström J. Large-Scale Metabolomics and the Incidence of Cardiovascular Disease. J Am Heart Assoc 2023; 12:e026885. [PMID: 36645074 PMCID: PMC9939066 DOI: 10.1161/jaha.122.026885] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Background The study aimed to show the relationship between a large number of circulating metabolites and subsequent cardiovascular disease (CVD) and subclinical markers of CVD in the general population. Methods and Results In 2278 individuals free from CVD in the EpiHealth study (aged 45-75 years, mean age 61 years, 50% women), 790 annotated nonxenobiotic metabolites were measured by mass spectroscopy (Metabolon). The same metabolites were measured in the PIVUS (Prospective Investigation of Vasculature in Uppsala Seniors) study (n=603, all aged 80 years, 50% women), in which cardiac and carotid artery pathologies were evaluated by ultrasound. During a median follow-up of 8.6 years, 107 individuals experienced a CVD (fatal or nonfatal myocardial infarction, stroke, or heart failure) in EpiHealth. Using a false discovery rate of 0.05 for age- and sex-adjusted analyses and P<0.05 for adjustment for traditional CVD risk factors, 37 metabolites were significantly related to incident CVD. These metabolites belonged to multiple biochemical classes, such as amino acids, lipids, and nucleotides. Top findings were dimethylglycine and N-acetylmethionine. A lasso selection of 5 metabolites improved discrimination when added on top of traditional CVD risk factors (+4.0%, P=0.0054). Thirty-five of the 37 metabolites were related to subclinical markers of CVD evaluated in the PIVUS study. The metabolite 1-carboxyethyltyrosine was associated with left atrial diameter as well as inversely related to both ejection fraction and the echogenicity of the carotid artery. Conclusions Several metabolites were discovered to be associated with future CVD, as well as with subclinical markers of CVD. A selection of metabolites improved discrimination when added on top of CVD risk factors.
Collapse
Affiliation(s)
- Lars Lind
- Department of Medical SciencesUppsala UniversityUppsalaSweden
| | - Tove Fall
- Department of Medical SciencesUppsala UniversityUppsalaSweden
| | - Johan Ärnlöv
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Science and SocietyKarolinska InstitutetHuddingeSweden,School of Health and Social StudiesDalarna UniversityFalunSweden
| | - Sölve Elmståhl
- Division of Geriatric Medicine, Department of Clinical Sciences in MalmöLund UniversityMalmöSweden
| | - Johan Sundström
- Department of Medical SciencesUppsala UniversityUppsalaSweden
| |
Collapse
|
35
|
Zhang M, Brady TM, Buckley JP, Appel LJ, Hong X, Wang G, Liang L, Wang X, Mueller NT. Metabolome-Wide Association Study of Cord Blood Metabolites With Blood Pressure in Childhood and Adolescence. Hypertension 2022; 79:2806-2820. [PMID: 36111548 PMCID: PMC9649875 DOI: 10.1161/hypertensionaha.122.20139] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 08/28/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND No studies have examined whether the cord blood metabolome-a reflection of in utero metabolism-influences blood pressure (BP) in children. OBJECTIVES To examine prospective associations of cord blood metabolites with systolic BP (SBP), diastolic BP (DBP), and risk of elevated BP in childhood and adolescence. METHODS In the Boston Birth Cohort, we measured metabolites in cord blood plasma, and SBP and DBP at clinic visits between 3 and 18 years. We examined associations of cord metabolites with SBP and DBP percentiles using linear mixed models and with elevated BP using mixed-effects Poisson regression. RESULTS Our study included 902 mother-child dyads (60% Black, 23% Hispanic, 45% female). Children were followed for a median of 9.2 (interquartile range, 6.7-11.7) years, and the median number of BP observations per child was 7 (interquartile range, 4-11). After false discovery rate correction, 3 metabolites were associated with SBP, 96 with DBP, and 24 with elevated BP; 2 metabolites (1-methylnicotinamide, dimethylguanidino valeric acid) were associated with all 3 outcomes, and 21 metabolites were associated with both DBP and elevated BP. After multivariable adjustment, 48 metabolites remained significantly associated with DBP. Metabolites that showed the strongest associations with SBP, DBP, and elevated BP included nucleotides (eg, xanthosine, hypoxanthine, xanthine) and acylcarnitines (eg, C6 and C7 carnitines), which represent fatty acid oxidation and purine metabolism pathways. CONCLUSIONS In our urban and predominantly racial/ethnic minority cohort, we provide evidence that metabolomic alterations in utero, in particular, acylcarnitine- and purine-metabolism metabolites, may be involved in the early life origins of hypertension.
Collapse
Affiliation(s)
- Mingyu Zhang
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD
| | - Tammy M Brady
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jessie P Buckley
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Lawrence J Appel
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD
- Division of General Internal Medicine, Johns Hopkins School of Medicine, Baltimore, MD
| | - Xiumei Hong
- Center on the Early Life Origins of Disease, Johns Hopkins University, Baltimore, MD
- Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Guoying Wang
- Center on the Early Life Origins of Disease, Johns Hopkins University, Baltimore, MD
- Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Liming Liang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Xiaobin Wang
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
- Center on the Early Life Origins of Disease, Johns Hopkins University, Baltimore, MD
- Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Noel T Mueller
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
36
|
Ghaeli T, Karimi B, Kojouri GA, Dehkordi RR, Ahadi AM. The Influence of Age and Gender on the Serum XOR Activity, Leukocyte Gene Expression of XOR and MPO, and Biochemical Parameters in Newborn Foals. J Equine Vet Sci 2022; 119:104134. [PMID: 36202292 DOI: 10.1016/j.jevs.2022.104134] [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: 08/10/2022] [Revised: 09/23/2022] [Accepted: 09/30/2022] [Indexed: 11/21/2022]
Abstract
One of the initial responses of the host's innate immunity of newborns against pathogens is the use of oxidative enzymes. This study aimed to evaluate changes in serum xanthine oxidoreductase (XOR) activity, the leukocyte myeloperoxidase (MPO) and XOR genes expression, and some biochemical parameters in healthy Darehshuri newborn foals up to 60 days of life. Blood samples were collected from 16 foals at 1, 7, 15, 30, and 60 days and used for detecting XOR activity, biochemical parameters, and also gene expression by real-time RT-PCR. High activity of XOR was observed at birth, explained by physiologic hypoxia during the birth without sex difference. The significant decrease in XOR activity during the following days is probably related to the decreased levels of substrate and feedback inhibition of XOR by uric acid. No correlations were found between XOR activity and uric acid. A positive correlation was observed between XOR mRNA and serum XOR activity in 15 days. The results also indicate higher levels of MPO gene expression at 30 days, which may be associated with their capacity for neutrophil phagocytosis. The concentrations of creatinine, total protein, and albumin were higher at birth, whereas uric acid level was lower (P < 0.05). It can be concluded that XOR activity decreases with age and there is no significant change in its gene expression and MPO expression increases with age and is sex-dependent. There is an influence of age on XOR activity, leukocyte expression of MPO, and biochemical parameters in healthy newborn foals up to 60 days of life.
Collapse
Affiliation(s)
- Taranom Ghaeli
- Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | - Behnaz Karimi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran.
| | - Gholam Ali Kojouri
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | | | - Ali Mohammad Ahadi
- Department of Genetic, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| |
Collapse
|
37
|
Allegrini S, Garcia-Gil M, Pesi R, Camici M, Tozzi MG. The Good, the Bad and the New about Uric Acid in Cancer. Cancers (Basel) 2022; 14:cancers14194959. [PMID: 36230882 PMCID: PMC9561999 DOI: 10.3390/cancers14194959] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/30/2022] [Accepted: 10/06/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The concentration of uric acid in blood is sex-, age- and diet-dependent and is maintained close to its maximal solubility, indicating that it plays some important role. Indeed, it has been demonstrated that, at physiological concentrations, uric acid is a powerful antioxidant and is a scavenger of singlet oxygen and radicals. At high intracellular concentration, uric acid has been demonstrated to act as a pro-oxidant molecule. Recently, uric acid has been reported to affect the properties of several proteins involved in metabolic regulation and signaling, and the relationship between uric acid and cancer has been extensively investigated. In this review, we present the most recent results on the positive and negative effects played by uric acid in cancer and some new findings and hypotheses about the implication of this metabolite in the pathogenesis of several diseases such as metabolic syndrome, diabetes, and inflammation, thus favoring the development of cancer. Abstract Uric acid is the final product of purine catabolism in man and apes. The serum concentration of uric acid is sex-, age- and diet-dependent and is maintained close to its maximal solubility, indicating that it plays some important role. Indeed, it has been demonstrated that, at physiological concentrations, uric acid is a powerful antioxidant, while at high intracellular concentrations, it is a pro-oxidant molecule. In this review, we describe the possible causes of uric acid accumulation or depletion and some of the metabolic and regulatory pathways it may impact. Particular attention has been given to fructose, which, because of the complex correlation between carbohydrate and nucleotide metabolism, causes uric acid accumulation. We also present recent results on the positive and negative effects played by uric acid in cancer and some new findings and hypotheses about the implication of this metabolite in a variety of signaling pathways, which can play a role in the pathogenesis of diseases such as metabolic syndrome, diabetes, and inflammation, thus favoring the development of cancer. The loss of uricase in Homo sapiens and great apes, although exposing these species to the potentially adverse effects of uric acid, appears to be associated with evolutionary advantages.
Collapse
Affiliation(s)
- Simone Allegrini
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, Università di Pisa, 56126 Pisa, Italy
- CISUP, Centro per L’Integrazione della Strumentazione dell’Università di Pisa, 56127 Pisa, Italy
- Correspondence:
| | - Mercedes Garcia-Gil
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, Università di Pisa, 56126 Pisa, Italy
- CISUP, Centro per L’Integrazione della Strumentazione dell’Università di Pisa, 56127 Pisa, Italy
- Unità di Fisiologia Generale, Dipartimento di Biologia, Università di Pisa, Via San Zeno 31, 56127 Pisa, Italy
| | - Rossana Pesi
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy
| | - Marcella Camici
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy
| | - Maria Grazia Tozzi
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy
| |
Collapse
|
38
|
Yang F, Hu T, Cui H. Serum urate and heart failure: a bidirectional Mendelian randomization study. Eur J Prev Cardiol 2022; 29:1570-1578. [PMID: 35578763 DOI: 10.1093/eurjpc/zwac100] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 11/13/2022]
Abstract
AIMS Observational studies indicate that serum urate level is associated with heart failure (HF). However, whether this association is causal remains controversial, due to confounding factors and reverse causality. We aim to evaluate the causal relationship of genetically predicted serum urate level with HF. METHODS AND RESULTS A bidirectional Mendelian randomization (MR) study was performed. Instrumental variables were obtained from the largest genome-wide association studies of serum urate (457 690 individuals) to date. We obtained summary statistics of HF from HERMES consortium (47 309 cases; 930 014 controls), the FinnGen study (13 087 cases; 195 091 controls), and the UK Biobank study (1088 cases; 360 106 controls). Inverse-variance-weighted method was applied to obtain MR estimates and other statistical methods were conducted in the sensitivity analyses. The reverse MR analysis was performed to evaluate the effect of HF on serum urate levels. Genetically determined serum urate level was associated with HF [odds ratio (OR), 1.07; 95% confidence interval (CI), 1.03-1.10; P = 8.6×10-5]. The main results kept robust in the most sensitivity analyses. The association pattern remained for the HF in FinnGen (OR, 1.10; 95% CI, 1.03-1.19; P = 0.008) and the combined results of three data sources (OR, 1.08; 95% CI, 1.04-1.13; P < 0.001). No consistent evidence was found for the causal effect of HF on serum urate levels. CONCLUSION We provide consistent evidence for the causal effect of genetically predicted serum urate level on HF, but not the reverse effect of HF. Urate-lowering therapy may be of cardiovascular benefit in the prevention of HF.
Collapse
Affiliation(s)
- Fangkun Yang
- Department of Cardiology, Ningbo Hospital of Zhejiang University (Ningbo First Hospital), School of Medicine, Zhejiang University, Ningbo, China
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou, China
- Cardiology Centre, Ningbo First Hospital, Ningbo University, 59 Liuting Road, Ningbo 315010, China
| | - Teng Hu
- School of Medicine, Ningbo University, Ningbo, China
| | - Hanbin Cui
- Cardiology Centre, Ningbo First Hospital, Ningbo University, 59 Liuting Road, Ningbo 315010, China
| |
Collapse
|
39
|
Rendić SP, Crouch RD, Guengerich FP. Roles of selected non-P450 human oxidoreductase enzymes in protective and toxic effects of chemicals: review and compilation of reactions. Arch Toxicol 2022; 96:2145-2246. [PMID: 35648190 PMCID: PMC9159052 DOI: 10.1007/s00204-022-03304-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/26/2022] [Indexed: 12/17/2022]
Abstract
This is an overview of the metabolic reactions of drugs, natural products, physiological compounds, and other (general) chemicals catalyzed by flavin monooxygenase (FMO), monoamine oxidase (MAO), NAD(P)H quinone oxidoreductase (NQO), and molybdenum hydroxylase enzymes (aldehyde oxidase (AOX) and xanthine oxidoreductase (XOR)), including roles as substrates, inducers, and inhibitors of the enzymes. The metabolism and bioactivation of selected examples of each group (i.e., drugs, "general chemicals," natural products, and physiological compounds) are discussed. We identified a higher fraction of bioactivation reactions for FMO enzymes compared to other enzymes, predominately involving drugs and general chemicals. With MAO enzymes, physiological compounds predominate as substrates, and some products lead to unwanted side effects or illness. AOX and XOR enzymes are molybdenum hydroxylases that catalyze the oxidation of various heteroaromatic rings and aldehydes and the reduction of a number of different functional groups. While neither of these two enzymes contributes substantially to the metabolism of currently marketed drugs, AOX has become a frequently encountered route of metabolism among drug discovery programs in the past 10-15 years. XOR has even less of a role in the metabolism of clinical drugs and preclinical drug candidates than AOX, likely due to narrower substrate specificity.
Collapse
Affiliation(s)
| | - Rachel D Crouch
- College of Pharmacy and Health Sciences, Lipscomb University, Nashville, TN, 37204, USA
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232-0146, USA
| |
Collapse
|
40
|
Deng W, Zheng Z, Chen Y, Yang M, Yan J, Li W, Zeng J, Xie J, Gong S, Zeng H. Deficiency of GntR Family Regulator MSMEG_5174 Promotes Mycobacterium smegmatis Resistance to Aminoglycosides via Manipulating Purine Metabolism. Front Microbiol 2022; 13:919538. [PMID: 35898907 PMCID: PMC9309504 DOI: 10.3389/fmicb.2022.919538] [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: 04/13/2022] [Accepted: 06/20/2022] [Indexed: 11/30/2022] Open
Abstract
The increasing incidence of drug-resistant tuberculosis is still an emergency for global public health and a major obstacle to tuberculosis treatment. Therefore, deciphering the novel mechanisms of mycobacterial antibiotic resistance is crucial for combatting the rapid emergence of drug-resistant strains. In this study, we identified an unexpected role of Mycobacterium smegmatis GntR family transcriptional regulator MSMEG_5174 and its homologous gene Mycobacterium tuberculosis Rv1152 in aminoglycoside antibiotic resistance. Deficiency of MSMEG_5174 rendered Mycobacterium smegmatis highly resistant to aminoglycoside antibiotic treatment, and ectopic expression of Rv1152 in MSMEG_5174 mutants restored antibiotic-induced bacterial killing. We further demonstrated that MSMEG_5174 negatively regulates the expression of purine metabolism-related genes and the accumulation of purine metabolites. Moreover, overexpression of xanthine dehydrogenase MSMEG_0871 or xanthine treatment elicited a significant decrease in aminoglycoside antibiotic lethality for Mycobacterium smegmatis. Together, our findings revealed MSMEG_5174 as a metabolic regulator and hint toward unexplored crosstalk between purine metabolism and antibiotic resistance.
Collapse
Affiliation(s)
- Wanyan Deng
- The Joint Center for Infection and Immunity, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou, China
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Wanyan Deng,
| | - Zengzhang Zheng
- The Joint Center for Infection and Immunity, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou, China
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Yi Chen
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Maoyi Yang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Yan
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wu Li
- The Joint Center for Infection and Immunity, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou, China
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Jie Zeng
- Department of Respiratory Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
- Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Jianping Xie
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Sitang Gong
- The Joint Center for Infection and Immunity, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou, China
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
- Sitang Gong,
| | - Huasong Zeng
- The Joint Center for Infection and Immunity, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou, China
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
- Huasong Zeng,
| |
Collapse
|
41
|
Chen MM, Meng LH. The double faced role of xanthine oxidoreductase in cancer. Acta Pharmacol Sin 2022; 43:1623-1632. [PMID: 34811515 PMCID: PMC9253144 DOI: 10.1038/s41401-021-00800-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/19/2021] [Indexed: 01/02/2023] Open
Abstract
Xanthine oxidoreductase (XOR) is a critical, rate-limiting enzyme that controls the last two steps of purine catabolism by converting hypoxanthine to xanthine and xanthine to uric acid. It also produces reactive oxygen species (ROS) during the catalytic process. The enzyme is generally recognized as a drug target for the therapy of gout and hyperuricemia. The catalytic products uric acid and ROS act as antioxidants or oxidants, respectively, and are involved in pro/anti-inflammatory actions, which are associated with various disease manifestations, including metabolic syndrome, ischemia reperfusion injury, cardiovascular disorders, and cancer. Recently, extensive efforts have been devoted to understanding the paradoxical roles of XOR in tumor promotion. Here, we summarize the expression of XOR in different types of cancer and decipher the dual roles of XOR in cancer by its enzymatic or nonenzymatic activity to provide an updated understanding of the mechanistic function of XOR in cancer. We also discuss the potential to modulate XOR in cancer therapy.
Collapse
Affiliation(s)
- Man-man Chen
- grid.9227.e0000000119573309Division of Anti-tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Ling-hua Meng
- grid.9227.e0000000119573309Division of Anti-tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|
42
|
Association of Hyperuricemia with 10-Year Atherosclerotic Cardiovascular Disease Risk among Chinese Adults and Elders. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19116713. [PMID: 35682297 PMCID: PMC9180408 DOI: 10.3390/ijerph19116713] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/28/2022] [Accepted: 05/29/2022] [Indexed: 11/17/2022]
Abstract
The purpose of this cross-sectional study is to use a representative sample of the Chinese population and the atherosclerotic cardiovascular disease (ASCVD) risk assessment tool developed specifically for the Chinese to explore the prevalence of hyperuricemia (HUA) and the relationship between hyperuricemia and 10-year ASCVD risk in Chinese adults. Data was collected from the Chinese Physiological Constant and Health Condition survey. In this study, 12,988 subjects aged between 35 and 74 were selected by two-stage, cluster and random sampling. The sex-specific 10-year ASCVD risk scores equations, which were conducted by China-PAR project and specifically designed for Chinese adults, were used to assess the risk of developing ASCVD 10 years later. The ordinal regression model was used to explore the relationship between hyperuricemia and ASCVD risk. The total prevalence of hyperuricemia was 12.69%, and males’ prevalence was significantly higher than females (17.7% vs. 8.5%). Compared with people without hyperuricemia, the 10-year ASCVD risk scores of female with hyperuricemia increased significantly, but no significant increased happened in male. The ordinal regression model indicated that hyperuricemia subjects were 1.3 (males, 95% CI: 1.11–1.52) and 4.34 (females, 95% CI: 3.16–5.91) times more likely to increase their ASCVD risk levels than those without hyperuricemia. In conclusion, Hyperuricemia is prevalent among Chinese adults. In both genders, hyperuricemia was related with higher risk of 10-year ASCVD, and the relationship is much stronger in females than in males. Thus, sex-specific serum uric acid management and intervention strategies should be done in the prevention and control of cardiovascular events.
Collapse
|
43
|
Kosaki K, Kumamoto S, Tokinoya K, Yoshida Y, Sugaya T, Murase T, Akari S, Nakamura T, Nabekura Y, Takekoshi K, Maeda S. Xanthine Oxidoreductase Activity in Marathon Runners: Potential Implications for Marathon-Induced Acute Kidney Injury. J Appl Physiol (1985) 2022; 133:1-10. [DOI: 10.1152/japplphysiol.00669.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Excess activation of circulating xanthine oxidoreductase (XOR) may contribute to the pathogenesis of widespread remote organ injury, including kidney injury. The purpose of this study was to determine the acute impact of marathon running on plasma XOR activity and to examine whether plasma XOR activity is associated with marathon-induced elevations in biomarkers of acute kidney injury (AKI). Twenty-three young men (aged 20-25 years) who participated in the 38th Tsukuba Marathon were included. Blood and urine samples were collected before, immediately, 2 h (only blood sample), and 24 h after a full marathon run. Plasma XOR activity was evaluated using a highly sensitive assay utilizing a combination of [13C2,15N2] xanthine and liquid chromatography-triple quadrupole mass spectrometry. The levels of several AKI biomarkers, such as serum creatinine and urinary liver-type fatty acid-binding protein (L-FABP) were measured in each participant. Marathon running caused a transient elevation in plasma XOR activity and levels of purine degradation products (hypoxanthine, xanthine, and uric acid) as well as serum creatinine, urinary albumin, and urinary L-FABP levels. Immediately after the marathon, individual relative changes in plasma XOR activity were independently correlated with corresponding changes in serum creatinine and urinary L-FABP levels. In addition, the magnitude of marathon-induced elevation in plasma XOR activity and levels of purine degradation products were higher in individuals who developed AKI. These findings collectively suggest that marathon running substantially influences the purine metabolism pathway including XOR activity. Moreover, activated circulating XOR can be partly associated with elevated biomarkers of AKI after marathon running.
Collapse
Affiliation(s)
- Keisei Kosaki
- Faculty of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
| | - Shota Kumamoto
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Katsuyuki Tokinoya
- Department of Clinical Laboratory, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
- Graduate School of Human Health Science, Tokyo Metropolitan University, Tokyo, Japan
- Japan Society for the Promotion of Science, Tokyo, Japan
| | - Yasuko Yoshida
- Department of Clinical Laboratory, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
- Department of Clinical Laboratory Science, Faculty of Health Sciences, Tsukuba International University, Ibaraki, Japan
| | - Takeshi Sugaya
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kanagawa, Japan
| | | | - Seigo Akari
- Sanwa Kagaku Kenkyusho Co., Ltd., Mie, Japan
| | | | - Yoshiharu Nabekura
- Faculty of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
| | - Kazuhiro Takekoshi
- Department of Clinical Laboratory, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Seiji Maeda
- Faculty of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
- Faculty of Sport Sciences, Waseda University, Saitama, Japan
| |
Collapse
|
44
|
The Intestinal Redox System and Its Significance in Chemotherapy-Induced Intestinal Mucositis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7255497. [PMID: 35585883 PMCID: PMC9110227 DOI: 10.1155/2022/7255497] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/04/2022] [Accepted: 04/09/2022] [Indexed: 12/12/2022]
Abstract
Chemotherapy-induced intestinal mucositis (CIM) is a significant dose-limiting adverse reaction brought on by the cancer treatment. Multiple studies reported that reactive oxygen species (ROS) is rapidly produced during the initial stages of chemotherapy, when the drugs elicit direct damage to intestinal mucosal cells, which, in turn, results in necrosis, mitochondrial dysfunction, and ROS production. However, the mechanism behind the intestinal redox system-based induction of intestinal mucosal injury and necrosis of CIM is still undetermined. In this article, we summarized relevant information regarding the intestinal redox system, including the composition and regulation of redox enzymes, ROS generation, and its regulation in the intestine. We innovatively proposed the intestinal redox “Tai Chi” theory and revealed its significance in the pathogenesis of CIM. We also conducted an extensive review of the English language-based literatures involving oxidative stress (OS) and its involvement in the pathological mechanisms of CIM. From the date of inception till July 31, 2021, 51 related articles were selected. Based on our analysis of these articles, only five chemotherapeutic drugs, namely, MTX, 5-FU, cisplatin, CPT-11, and oxaliplatin were shown to trigger the ROS-based pathological mechanisms of CIM. We also discussed the redox system-mediated modulation of CIM pathogenesis via elaboration of the relationship between chemotherapeutic drugs and the redox system. It is our belief that this overview of the intestinal redox system and its role in CIM pathogenesis will greatly enhance research direction and improve CIM management in the future.
Collapse
|
45
|
Yang F, Lu Y, Chen S, Wang K, Hu T, Cui H. Sex-specific effect of serum urate levels on coronary heart disease and myocardial infarction prevention: A Mendelian randomization study. Nutr Metab Cardiovasc Dis 2022; 32:1266-1274. [PMID: 35197211 DOI: 10.1016/j.numecd.2022.01.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND AIMS Observational studies have examined serum urate levels in relation to coronary heart disease (CHD) and myocardial infarction (MI). Whether these associations are causal remains controversial, due to confounding factors and reverse causality. We aim to investigate the causality of these associations using Mendelian randomization method. METHODS AND RESULTS Instrumental variables were obtained from the largest genome-wide association studies of serum urate (457,690 individuals) to date. Summary statistics were from CARDIoGRAMplusC4D consortium (60,801 CHD cases; 43,676 MI cases), FinnGen (21,012 CHD cases; 12,801 MI cases), UK Biobank (10,157 CHD cases; 7018 MI cases), and Biobank Japan (29,319 CHD cases). Inverse-variance weighted method was applied as the main results. Other statistical methods and reverse MR analysis were conducted in the supplementary analyses. Elevated genetically determined serum urate levels were associated with increased risks of CHD and MI. The association pattern remained for the datasets in FinnGen, the combined results of three independent data sources (CHD: odds ratio (OR), 1.10; 95%CI, 1.06-1.15; p = 4.2 × 10-6; MI: OR, 1.12; 95%CI, 1.07-1.18; p = 2.7 × 10-6), and East Asian population. Interestingly, sex-specific subgroup analyses revealed that these associations kept in men only, but not among women in individuals of European ancestry. No consistent evidence was found for the causal effect of CHD or MI on serum urate levels. CONCLUSION We provide consistent evidence for the causal effect of genetically predicted serum urate levels on CHD and MI, but not the reverse effect. Urate-lowering therapy may be of cardiovascular benefit in the prevention of CHD and MI, especially for men.
Collapse
Affiliation(s)
- Fangkun Yang
- Department of Cardiology, Ningbo Hospital of Zhejiang University (Ningbo First Hospital), School of Medicine, Zhejiang University, Ningbo, Zhejiang, China
| | - Yunlong Lu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Songzan Chen
- School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kai Wang
- Eye Center of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Teng Hu
- School of Medicine, Ningbo University, Ningbo, China
| | - Hanbin Cui
- Cardiology Center, Ningbo First Hospital, Ningbo University, Ningbo, Zhejiang, China.
| |
Collapse
|
46
|
Nishizawa H, Maeda N, Shimomura I. Impact of hyperuricemia on chronic kidney disease and atherosclerotic cardiovascular disease. Hypertens Res 2022; 45:635-640. [PMID: 35046512 DOI: 10.1038/s41440-021-00840-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/30/2021] [Indexed: 11/09/2022]
Abstract
Hyperuricemia is caused by reduced renal/extrarenal excretion and overproduction of uric acid. It is affected by genetic predisposition related to uric acid transporters and by visceral fat accumulation due to overnutrition. The typical symptomatic complication of hyperuricemia is gout caused by monosodium urate crystals. Accumulated evidence from epidemiological studies suggests that hyperuricemia is also a risk factor for hypertension, chronic kidney disease (CKD) and atherosclerotic cardiovascular disease (CVD). However, it remains to be determined whether urate-lowering therapy for asymptomatic patients with hyperuricemia is effective in preventing CKD or CVD progression. This mini review focuses mainly on recent papers investigating the relationship between hyperuricemia and CKD or CVD and studies of urate-lowering therapy. Accumulated studies have proposed mechanisms of renal damage and atherosclerosis in hyperuricemia, including inflammasome activation, decreased nitric oxide bioavailability and oxidative stress induced by uric acid, urate crystals and xanthine oxidoreductase (XOR)-mediated reactive oxygen species. Since patients with hyperuricemia are a heterogeneous population with complex pathologies, it may be important to assess whether an outcome is the result of decreasing serum uric acid levels or an inhibitory effect on XOR. To clarify the impact of hyperuricemia on CKD and CVD progression, high-quality and detailed clinical and basic science studies of hyperuricemia and purine metabolism are needed.
Collapse
Affiliation(s)
- Hitoshi Nishizawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, 2-2, Yamada-oka, Suita, Osaka, 565-0871, Japan.
| | - Norikazu Maeda
- Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, 2-2, Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, 2-2, Yamada-oka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
47
|
Xanthine oxidoreductase activity in platelet-poor and rich plasma as a oxidative stress indicator in patients required renal replacement therapy. BMC Nephrol 2022; 23:35. [PMID: 35042470 PMCID: PMC8764817 DOI: 10.1186/s12882-021-02649-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/21/2021] [Indexed: 11/10/2022] Open
Abstract
Background Xanthine oxidoreductase (XOR) is a hydroxylase enzyme involved in the metabolism of purines. XOR activity can vary: the homodimer protein can be converted into two different isoforms XD (antioxidant) and XO (prooxidant). Oxidative stress and inflammation that accompanying chronic kidney disease (CKD), dialysis, and kidney transplantation, resulted in platelet activation. Present study aimed to determine the influence of applied renal replacement therapy on xanthine oxidoreductase and its isoforms activity. Materials and Methods The study group consisted of 117 patients, divided into 4 groups: hemodialysis - 30 patients, peritoneal dialysis - 30 patients, kidney transplant patients - 27 and conservative treatment - 30 patients. The control group consisted of 30 healthy volunteers. Results Significant differences were found in XOR activity in platelet-poor plasma (PPP) within the groups studied (p = 0.001). There was a relationship between the type of renal replacement therapy of all oxidoreductase isoforms in PPP (p < 0.001 all isoforms) and XD (p = 0.008), XO (p < 0.001) in platelet rich-plasma (PRP). A relationship was observed between the activity of all oxidoreductase isoforms in PPP and PRP, and the type of renal replacement therapy and the duration of dialysis and the age of patients. The cause of chronic kidney disease was also reflected differences in XD and XO activity in PPP. Conclusions The type of renal replacement therapy used in CKD patients, age of patients, duration of dialysis, CKD causes, and stage of progression significantly affect the activity of XOR and its isoforms.
Collapse
|
48
|
Polito L, Bortolotti M, Battelli MG, Bolognesi A. Xanthine oxidoreductase: A leading actor in cardiovascular disease drama. Redox Biol 2021; 48:102195. [PMID: 34844041 PMCID: PMC8636850 DOI: 10.1016/j.redox.2021.102195] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/19/2021] [Accepted: 11/19/2021] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular diseases (CVD) are the leading cause of global mortality and their pathogenesis lies mainly in the atherosclerotic process. There are close connections linking oxidative stress and inflammation to endothelial dysfunction, atherosclerosis and, consequently, to CVD. This review focuses on the role of xanthine oxidoreductase (XOR) and its products on the development of chronic inflammation and oxidative stress, responsible for atheromatous plaque formation. Evidence is reported that an excessive level of XOR products favors inflammatory response and plaque development, thereby promoting major cardiovascular risk factors. Also, the relationship between hyperuricemia and hypertension as well as between XOR activity and CVD is confirmed. In spite of the increasing number of clinical studies investigating the output of cardiovascular patients treated with urate-lowering therapies (including uricosuric drugs, XOR inhibitors and recombinant uricase) the results are still uncertain. The inhibition of XOR activity appears more promising than just the control of uricemia level in preventing cardiovascular events, possibly because it also reduces the intracellular accumulation of urate, as well as the production of reactive oxygen species. However, XOR inhibition also reduces the availability of the multifaced mediator nitric oxide and, at present, can be recommended only in hyperuricemic patients.
Collapse
Affiliation(s)
- Letizia Polito
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Via San Giacomo 14, 40126, Bologna, Italy.
| | - Massimo Bortolotti
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Via San Giacomo 14, 40126, Bologna, Italy.
| | - Maria Giulia Battelli
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Via San Giacomo 14, 40126, Bologna, Italy.
| | - Andrea Bolognesi
- Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum, University of Bologna, Via San Giacomo 14, 40126, Bologna, Italy.
| |
Collapse
|
49
|
Mineral Micronutrients in Asthma. Nutrients 2021; 13:nu13114001. [PMID: 34836256 PMCID: PMC8625329 DOI: 10.3390/nu13114001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 01/01/2023] Open
Abstract
Asthma represents one of the most common medical issues in the modern world. It is a chronic inflammatory disease characterized by persistent inflammation of the airways and disturbances in redox status, leading to hyperresponsiveness of bronchi and airway obstruction. Apart from classical risk factors such as air pollution, family history, allergies, or obesity, disturbances of the levels of micronutrients lead to impairments in the defense mechanisms of the affected organism against oxidative stress and proinflammatory stimuli. In the present review, the impact of micronutrients on the prevalence, severity, and possible risk factors of asthma is discussed. Although the influence of classical micronutrients such as selenium, copper, or zinc are well known, the effects of those such as iodine or manganese are only rarely mentioned. As a consequence, the aim of this paper is to demonstrate how disturbances in the levels of micronutrients and their supplementation might affect the course of asthma.
Collapse
|
50
|
Hori M, Imamura T, Kataoka N, Nakamura M, Tanaka S, Onoda H, Ushijima R, Sobajima M, Fukuda N, Ueno H, Joho S, Kinugawa K. Urinary Isoxanthopterin in Heart Failure Patients. Circ Rep 2021; 3:654-659. [PMID: 34805605 PMCID: PMC8578125 DOI: 10.1253/circrep.cr-21-0112] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/02/2021] [Accepted: 09/04/2021] [Indexed: 11/09/2022] Open
Abstract
Background: The prognostic impact of urinary isoxanthopterin (U-IXP), a recently proposed marker of oxidative stress, in patients with heart failure remains unknown. Methods and Results: Patients who were admitted to our institute for decompensated heart failure were prospectively included in the study; U-IXP was measured on admission. The association between the U-IXP concentration and a composite primary outcome that included cardiovascular death and heart failure readmissions following the index discharge was investigated. In all, 42 patients (median age 78 years [interquartile range {IQR} 69-85 years]; 25 males) were included in the study. The median U-IXP concentration on admission was 0.58 μmol/g creatinine (Cre; IQR 0.35-0.95 μmol/g Cre). A higher U-IXP concentration was an independent predictor of the primary outcome adjusted for clinical potential confounders and was associated with a significantly higher cumulative incidence of the primary outcome (71% vs. 16%, P=0.001) at a cut-off of 0.93 μmol/g Cre. Conclusions: U-IXP on admission was associated with cardiovascular death or heart failure readmission following the index discharge in patients with decompensated heart failure. The clinical implication of aggressive interventions to normalize U-IXP and the detailed prognostic mechanism of U-IXP in heart failure patients remain the next concerns.
Collapse
Affiliation(s)
- Masakazu Hori
- Second Department of Internal Medicine, University of Toyama Toyama Japan
| | - Teruhiko Imamura
- Second Department of Internal Medicine, University of Toyama Toyama Japan
| | - Naoya Kataoka
- Second Department of Internal Medicine, University of Toyama Toyama Japan
| | - Makiko Nakamura
- Second Department of Internal Medicine, University of Toyama Toyama Japan
| | - Shuhei Tanaka
- Second Department of Internal Medicine, University of Toyama Toyama Japan
| | - Hiroshi Onoda
- Second Department of Internal Medicine, University of Toyama Toyama Japan
| | - Ryuichi Ushijima
- Second Department of Internal Medicine, University of Toyama Toyama Japan
| | - Mitsuo Sobajima
- Second Department of Internal Medicine, University of Toyama Toyama Japan
| | - Nobuyuki Fukuda
- Second Department of Internal Medicine, University of Toyama Toyama Japan
| | - Hiroshi Ueno
- Second Department of Internal Medicine, University of Toyama Toyama Japan
| | - Shuji Joho
- Second Department of Internal Medicine, University of Toyama Toyama Japan
| | - Koichiro Kinugawa
- Second Department of Internal Medicine, University of Toyama Toyama Japan
| |
Collapse
|