1
|
Girigoswami K, Arunkumar R, Girigoswami A. Management of hypertension addressing hyperuricaemia: introduction of nano-based approaches. Ann Med 2024; 56:2352022. [PMID: 38753584 PMCID: PMC11100442 DOI: 10.1080/07853890.2024.2352022] [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: 01/30/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024] Open
Abstract
Uric acid (UA) levels in blood serum have been associated with hypertension, indicating a potential causal relationship between high serum UA levels and the progression of hypertension. Therefore, the reduction of serum UA level is considered a potential strategy for lowering and mitigating blood pressure. If an individual is at risk of developing or already manifesting elevated blood pressure, this intervention could be an integral part of a comprehensive treatment plan. By addressing hyperuricaemia, practitioners may subsidize the optimization of blood pressure regulation, which illustrates the importance of addressing UA levels as a valuable strategy within the broader context of hypertension management. In this analysis, we outlined the operational principles of effective xanthine oxidase inhibitors for the treatment of hyperuricaemia and hypertension, along with an exploration of the contribution of nanotechnology to this field.
Collapse
Affiliation(s)
- Koyeli Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai, India
| | - Radhakrishnan Arunkumar
- Department of Pharmacology, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai, India
| | - Agnishwar Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai, India
| |
Collapse
|
2
|
Zhu X, Zheng H, Zhang Z, Ma S, Feng Q, Wang J, Wu G, Ng HY. Cytotoxicity evaluation of organophosphorus flame retardants using electrochemical biosensors and elucidation of associated toxic mechanisms. WATER RESEARCH 2024; 265:122262. [PMID: 39167971 DOI: 10.1016/j.watres.2024.122262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 07/03/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
In recent years, organophosphorus flame retardants (OPFRs) have been widely used as substitutes for brominated flame retardants with excellent properties, and their initial toxicological effects on the water ecosystem and human health have gradually emerged. However, to date, research on the cytotoxicity and health risks of OPFRs is still limited. Therefore, this study aims to systematically explore the cytotoxic effects and toxic mechanisms of OPFRs on cells. Human liver cancer (HepG2) cells were adopted as an ideal model for toxicity evaluation due to their rapid growth and metabolism. This study proposes a sensitive electrochemical cell-based sensor constructed on a graphitized multi-walled carbon nanotube/ionic liquid/gold nanoparticle-modified electrode. The sensor was used to detect the cytotoxicity of tri(2-butylxyethyl) phosphate (TBEP), tributyl phosphate (TnBP), triphenyl phosphate (TPhP), tri(1,3-dichloro-2-propyl) phosphate (TDCIPP), tri(2-chloropropyl) phosphate (TCPP) and tri(2-chloroethyl) phosphate (TCEP) in the liquid medium, providing insight into their toxicity in water environments. The half-maximal inhibitory concentration (IC50) of TBEP, TnBP, TPhP, TDCIPP, TCPP and TCEP on HepG2 cells were 179.4, 194.9, 219.8, 339.4, 511.8 and 859.0 μM, respectively. Additionally, the cytotoxic mechanism of six OPFRs was discussed from the perspective of oxidative stress and apoptosis, and four indexes were correlated with toxicity. Furthermore, transcriptome sequencing was conducted, followed by a thorough analysis of the obtained sequencing results. This analysis demonstrated a significant enrichment of the p53 and PPAR pathways, both of which are closely associated with oxidative stress and apoptosis. This study presents a simplified and efficient technique for conducting in vitro toxicity studies on organophosphorus flame retardants in a water environment. Moreover, it establishes a scientific foundation for further investigation into the mechanisms of cytotoxicity associated with these compounds.
Collapse
Affiliation(s)
- Xiaolin Zhu
- School of Environment, Northeast Normal University, Changchun 130117, PR China
| | - Huizi Zheng
- School of Environment, Northeast Normal University, Changchun 130117, PR China
| | - Zhipeng Zhang
- School of Environment, Northeast Normal University, Changchun 130117, PR China
| | - Shuang Ma
- School of Environment, Northeast Normal University, Changchun 130117, PR China
| | - Qi Feng
- School of Environment, Northeast Normal University, Changchun 130117, PR China
| | - Jinsheng Wang
- Center for Water Research, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, 519087, China
| | - Guanlan Wu
- School of Environment, Northeast Normal University, Changchun 130117, PR China; Center for Water Research, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, 519087, China.
| | - How Yong Ng
- Center for Water Research, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, 519087, China; Department of Civil and Environmental Engineering, National University of Singapore, 1 Engineering Drive 2, 117576, Singapore.
| |
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
|
Chu L, Tian X, Chen S, Xia X, Xu Q, Zhang Y, Wu S, Wang A. Serum uric acid trajectories and the risk of cardiovascular disease: a longitudinal association and pathway analysis. Endocrine 2024:10.1007/s12020-024-04038-1. [PMID: 39365388 DOI: 10.1007/s12020-024-04038-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Age-specifically longitudinal associations and potential pathways between serum uric acid (SUA) and cardiovascular disease (CVD) remained unclear. This study aimed to explore SUA trajectories in different age populations and to determine their associations and potential pathways with incident CVD. METHODS This prospective cohort included 41,367 participants from the Kailuan study, including 30,938 participants aged <55 years and 10,419 participants aged ≥55. The SUA trajectories during year 2006-2012 were identified by latent class growth models. RESULTS Three SUA trajectories were identified in the overall, aged <55 and aged ≥55 years participants, as "low-stable" (51.9%, 54.4%, and 43.3%), "moderate-stable" (39.0%, 36.9%, and 45.6%), and "high-stable" (9.1%, 9.7%, and 11.1%), respectively. During a median follow-up of 6.75 years, incident CVD occurred in 2302 participants (5.56%). Overall, a high-stable trajectory was independently associated with a higher risk of CVD (hazard ratio [HR], 1.23; 95% [confidence interval], 1.06-1.42). Notably, the associations differed by age, a significant association was only observed in participants aged ≥55 years (HR, 1.29; 95% CI, 1.05-1.58), rather than those aged <55 years (HR, 1.08; 95% CI, 0.89-1.33). The addition of SUA trajectories to a baseline risk model for CVD improved the integrated discrimination improvement value (P < 0.05) and category-free net reclassification improvement value (P < 0.05). Bayesian network showed the conditional probability of high CVD risk associated with aging, elevated SUA trajectories, blood pressure, glucose, and inflammation was 15.5%. CONCLUSIONS High-stable SUA trajectories were independently associated with an elevated risk of CVD, which is mainly induced by hypertension, diabetes, and inflammation, especially in participants aged ≥55 years.
Collapse
Affiliation(s)
- Lulu Chu
- Department of Endocrinology, Kailuan Hospital, North China University of Science and Technology, Tangshan, China
| | - Xue Tian
- Department of Neuroepidemiology, Beijing Neurosurgical Institute, 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 Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Shuohua Chen
- Department of Cardiology, Kailuan Hospital, North China University of Science and Technology, Tangshan, China
| | - Xue Xia
- Department of Neuroepidemiology, Beijing Neurosurgical Institute, 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 Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China
| | - Qin Xu
- Department of Neuroepidemiology, Beijing Neurosurgical Institute, 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 Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China
| | - Yijun Zhang
- Department of Neuroepidemiology, Beijing Neurosurgical Institute, 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 Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Shouling Wu
- Department of Cardiology, Kailuan Hospital, North China University of Science and Technology, Tangshan, China.
| | - Anxin Wang
- Department of Neuroepidemiology, Beijing Neurosurgical Institute, 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 Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China.
| |
Collapse
|
5
|
Akopova O, Korkach Y, Sagach V. The effects of ecdysterone and enalapril on nitric oxide synthesis and the markers of oxidative stress in streptozotocin-induced diabetes in rats: a comparative study. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8089-8099. [PMID: 38789633 DOI: 10.1007/s00210-024-03154-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024]
Abstract
Cardiovascular functions in diabetes greatly depend on constitutive NOS (cNOS) activity. A comparative study of the effects of a steroid hormone ecdysterone and enalapril, an ACE inhibitor widely used to treat cardiac disorders on cNOS, inducible NOS (iNOS), xanthine oxidoreductase (XOR) activity, RNS, ROS, and lipid peroxidation in heart tissue in experimental diabetes was conducted. The rat model of diabetes was established by streptozotocin injection. NOS activity, NO2-, NO3-, uric acid, nitrosothiols, hydroperoxide, superoxide, and diene conjugate formation were studied spectrophotomerically. In diabetes, cNOS downregulation correlated with a dramatic fall of NO2- production and ~4.5-fold elevation of nitrosothiols, which agreed with a steep rise of iNOS activity, while NO3- remained close to control. Dramatic activation of XOR was observed, which correlated with the elevation of both superoxide production and nitrate reductase activity and resulted in strong lipid peroxidation. Ecdysterone and enalapril differently affected RNS metabolism. Ecdysterone moderately restored cNOS but strongly suppressed iNOS, which resulted in the reduction of NO3-, but full restoration of NO2- production. Enalapril better restored cNOS but less effectively suppressed iNOS, which promoted NO3- formation. Both drugs similarly inhibited XOR, which equally alleviated oxidative stress and lipid peroxidation. The synergistic action of iNOS and XOR was a plausible explanation for strong lipid peroxidation, abolished by the inhibition of iNOS and XOR by ecdysterone or enalapril. Complementary effects of ecdysterone and enalapril on cNOS, iNOS, and RNS are a promising basis for their combined use in the treatment of cardiovascular disorders caused by cNOS dysfunction in diabetes.
Collapse
Affiliation(s)
- Olga Akopova
- Stem cell laboratory, Bogomoletz Institute of Physiology, NAS of Ukraine, Kiev, Ukraine.
| | - Yulia Korkach
- Circulation department, Bogomoletz Institute of Physiology, NAS of Ukraine, Kiev, Ukraine
| | - Vadim Sagach
- Circulation department, Bogomoletz Institute of Physiology, NAS of Ukraine, Kiev, Ukraine
| |
Collapse
|
6
|
Lochhead JJ, Ronaldson PT, Davis TP. The role of oxidative stress in blood-brain barrier disruption during ischemic stroke: Antioxidants in clinical trials. Biochem Pharmacol 2024; 228:116186. [PMID: 38561092 PMCID: PMC11410550 DOI: 10.1016/j.bcp.2024.116186] [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: 01/22/2024] [Revised: 03/19/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Ischemic stroke is one of the leading causes of death and disability. Occlusion and reperfusion of cerebral blood vessels (i.e., ischemia/reperfusion (I/R) injury) generates reactive oxygen species (ROS) that contribute to brain cell death and dysfunction of the blood-brain barrier (BBB) via oxidative stress. BBB disruption influences the pathogenesis of ischemic stroke by contributing to cerebral edema, hemorrhagic transformation, and extravasation of circulating neurotoxic proteins. An improved understanding of mechanisms for ROS-associated alterations in BBB function during ischemia/reperfusion (I/R) injury can lead to improved treatment paradigms for ischemic stroke. Unfortunately, progress in developing ROS targeted therapeutics that are effective for stroke treatment has been slow. Here, we review how ROS are produced in response to I/R injury, their effects on BBB integrity (i.e., tight junction protein complexes, transporters), and the utilization of antioxidant treatments in ischemic stroke clinical trials. Overall, knowledge in this area provides a strong translational framework for discovery of novel drugs for stroke and/or improved strategies to mitigate I/R injury in stroke patients.
Collapse
Affiliation(s)
- Jeffrey J Lochhead
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ 85724, USA.
| | - Patrick T Ronaldson
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | - Thomas P Davis
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| |
Collapse
|
7
|
Mayers J, Hofman B, Sobiech I, Kwesiga MP. Insights into the biocompatibility of biodegradable metallic molybdenum for cardiovascular applications-a critical review. Front Bioeng Biotechnol 2024; 12:1457553. [PMID: 39376544 PMCID: PMC11456422 DOI: 10.3389/fbioe.2024.1457553] [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: 07/01/2024] [Accepted: 09/02/2024] [Indexed: 10/09/2024] Open
Abstract
Atherosclerotic cardiovascular disease (ACD) is the leading cause of death worldwide. The gold standard of treatment is the implantation of a permanent stent implant that is often associated with complications such as thrombus formation, vascular neointimal response, and stent fracture, which altogether decrease the long-term safety and efficacy of the stent. Biodegradable metallic materials have become an attractive alternative because of the ability to facilitate a more physiological healing response while the metal degrades. Recently, Molybdenum (Mo) has been considered as a potential candidate due to its excellent mechanical and medical imaging properties. Moreover, the biomedical research studies performed to date have shown minimal adverse effects in vitro and in vivo. However, there are still concerns of toxicity at high doses, and the impact of the biochemical mechanisms of Mo on material performance especially in pathophysiological environments are yet to be explored. Mo is an essential co factor for enzymes such as xanthine oxidoreductase (XOR) that plays a critical role in vascular homeostasis and ACD progression. Herein, this review will focus on the biochemistry of Mo, its physiological and pathological effects with an emphasis on cardiovascular disease as well as the recent studies on Mo for cardiovascular applications and its advantages over other biodegradable metals. The limitations of Mo research studies will also be discussed and concluded with an outlook to move this revolutionary metallic biomaterial from the bench to the bedside.
Collapse
Affiliation(s)
- Janina Mayers
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI, United States
| | - Brianna Hofman
- Department of Cell and Molecular Biology, Grand Valley State University, Allendale, MI, United States
| | - Indie Sobiech
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI, United States
| | - Maria P. Kwesiga
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI, United States
| |
Collapse
|
8
|
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
|
9
|
Elias-Llumbet A, Sharmin R, Berg-Sorensen K, Schirhagl R, Mzyk A. The Interplay between Mechanoregulation and ROS in Heart Physiology, Disease, and Regeneration. Adv Healthc Mater 2024; 13:e2400952. [PMID: 38962858 DOI: 10.1002/adhm.202400952] [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: 03/13/2024] [Revised: 05/16/2024] [Indexed: 07/05/2024]
Abstract
Cardiovascular diseases are currently the most common cause of death in developed countries. Due to lifestyle and environmental factors, this problem is only expected to increase in the future. Reactive oxygen species (ROS) are a key player in the onset of cardiovascular diseases but also have important functions in healthy cardiac tissue. Here, the interplay between ROS generation and cardiac mechanical forces is shown, and the state of the art and a perspective on future directions are discussed. To this end, an overview of what is currently known regarding ROS and mechanosignaling at a subcellular level is first given. There the role of ROS in mechanosignaling as well as the interplay between both factors in specific organelles is emphasized. The consequences at a larger scale across the population of heart cells are then discussed. Subsequently, the roles of ROS in embryogenesis, pathogenesis, and aging are further discussed, exemplifying some aspects of mechanoregulation. Finally, different models that are currently in use are discussed to study the topics above.
Collapse
Affiliation(s)
- Arturo Elias-Llumbet
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, Groningen, 9713AW, The Netherlands
- Laboratory of Genomic of Germ Cells, Biomedical Sciences Institute, Faculty of Medicine, University of Chile, Independencia, Santiago, 1027, Chile
| | - Rokshana Sharmin
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, Groningen, 9713AW, The Netherlands
| | | | - Romana Schirhagl
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, Groningen, 9713AW, The Netherlands
| | - Aldona Mzyk
- DTU Health Tech, Ørsteds Plads Bldg 345C, Kongens Lyngby, 2800, Denmark
| |
Collapse
|
10
|
Xiao Y, Chen Y, Zuo X, Mutalifu K, Chen X, Liu K. The Effect of Tolvaptan on Metabolism and Electrolyte Homeostasis in Patients with Heart Failure: A Systematic Review and Meta-Analysis. Rev Cardiovasc Med 2024; 25:334. [PMID: 39355601 PMCID: PMC11440402 DOI: 10.31083/j.rcm2509334] [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: 05/09/2024] [Revised: 07/09/2024] [Accepted: 07/31/2024] [Indexed: 10/03/2024] Open
Abstract
Background This study aimed to investigate the effect of tolvaptan on metabolism and electrolyte homeostasis in patients with heart failure (HF). Methods Literature databases, such as PubMed, EMBASE, the Cochrane Library, China National Knowledge Infrastructure, VIP, and WanFang Data, were systematically searched for relevant trials from inception to November 4, 2023. We used the fixed effect model to combine the effect sizes and used I2 to test heterogeneity. Funnel plots were plotted to assess publication bias. Results 16 studies were eligible for further analysis. No significant differences were identified in the incidence of hyperuricemia between the tolvaptan group and the placebo group (odds ratio (OR) = 1.23, 95% confidence interval (CI) = 0.97 to 1.55, p = 0.09). Tolvaptan decreased the levels of blood uric acid compared to traditional diuretics (mean difference (MD) = -82.8, 95% CI = -96.48 to -69.13, p < 0.00001). There was no significant difference in hypernatremia (OR = 1.62, 95% CI = 0.66 to 3.96, p = 0.29) and hyperkalemia (OR = 1.17, 95% CI = 0.93 to 1.48, p = 0.18) between the tolvaptan and control groups. Conclusions Tolvaptan reduced the level of blood uric acid compared to conventional diuretics, and could be used as a substitute for traditional diuretics for HF patients with a high risk of gout.
Collapse
Affiliation(s)
- Yao Xiao
- Department of Cardiology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Yue Chen
- Department of Clinical Medicine, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
- Department of Pharmacy, West China Tianfu Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Xianghao Zuo
- Department of Cardiology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Kadireya Mutalifu
- West China School of Medicine/West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Xiaoping Chen
- Department of Cardiology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Kai Liu
- Department of Cardiology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
| |
Collapse
|
11
|
Yokota T, Kinugawa S, Fukushima A, Okumura T, Murohara T, Tsutsui H. Efficacy and safety of the urate-lowering agent febuxostat in chronic heart failure patients with hyperuricemia: results from the LEAF-CHF study. Heart Vessels 2024:10.1007/s00380-024-02448-9. [PMID: 39158751 DOI: 10.1007/s00380-024-02448-9] [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: 01/16/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024]
Abstract
Hyperuricemia is an independent predictor of mortality in patients with chronic heart failure (CHF). To determine whether febuxostat, a urate-lowering agent, may improve clinical outcomes in CHF patients, we conducted a multicenter, prospective, randomized, open-label, blinded endpoint study with a treatment period of 24 weeks. We randomly assigned Japanese outpatients diagnosed with both CHF with reduced left ventricular ejection fraction (LVEF < 40%) and asymptomatic hyperuricemia (serum uric acid [UA] levels > 7.0 mg/dl and < 10.0 mg/dl) to either a febuxostat group (n = 51) or a control group (n = 50). The primary efficacy endpoint was the change in log-transformed plasma B-type natriuretic peptide (BNP) levels from baseline to week 24 (or at discontinuation). The secondary efficacy endpoints were the changes in LV systolic or diastolic function evaluated by echocardiography, New York Heart Association (NYHA) class, hemoglobin, and estimated glomerular filtration rate from baseline to week 24, and the change in log-transformed plasma BNP levels or serum UA levels from baseline to weeks 4, 8, 12, 16 and 20 (BNP) or weeks 4, 8, 12, 16, 20 and 24 (serum UA). The primary safety endpoints were occurrence of all-cause death or major cardiovascular events. The mean age of participants was 70 years; 14% were female. The febuxostat group and the control group did not differ with respect to the primary efficacy endpoint (p = 0.13), although the decrease in log-transformed plasma BNP levels from baseline to each of weeks 4, 8, 12, 16 and 20 was greater in the febuxostat group. There were no significant differences between the two groups in the primary safety endpoints or the secondary efficacy endpoints except reduced serum UA levels in the febuxostat group. Febuxostat did not reduce plasma BNP levels at week 24 in patients with CHF, but it appeared safe with no increase in major cardiovascular events and all-cause or cardiovascular mortality.
Collapse
Affiliation(s)
- Takashi Yokota
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.
- Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Kita-14 Nishi-5, Kita-ku, Sapporo, 060-8648, Japan.
| | - Shintaro Kinugawa
- Department Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Faculty of Medical Sciences, Research Institute of Angiocardiology, Kyushu University, Fukuoka, Japan
| | - Arata Fukushima
- Department of Cardiovascular Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takahiro Okumura
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroyuki Tsutsui
- School of Medicine and Graduate School, International University of Health and Welfare, Okawa, Japan
| |
Collapse
|
12
|
Piani F, Baschino S, Agnoletti D, Calandrini L, Degli Esposti D, Di Micoli A, Falcone R, Fiorini G, Ianniello E, Mauloni P, Ventura F, Veronesi M, Johnson RJ, Borghi C. Serum uric acid to eGFR ratio correlates with adverse outcomes in elderly hospitalized for acute heart failure. Int J Cardiol 2024; 409:132160. [PMID: 38740335 DOI: 10.1016/j.ijcard.2024.132160] [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: 02/20/2024] [Revised: 05/01/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND Serum uric acid (SUA) is a known biomarker of severity in acute heart failure (AHF), reflecting the intricate interplay between cardiovascular and metabolic dysfunction. Since SUA can increase in response to worsening kidney function, and subjects with AHF often have cardiorenal syndrome or are on diuretic therapy, we tested whether the ratio of SUA to eGFR might provide prognostic value in elderly hospitalized for AHF. METHODS The BOTERO-AHF Study (BOlogna study of Therapies, Epidemiology and Radiodiagnostic Outcomes in Acute Heart Failure patients) included 293 patients admitted for AHF who were consecutively enrolled from January 2020 onwards. We compared the baseline characteristics of participants who had a composite outcome (CO) (n = 203) of death or re-hospitalization for AHF within 12 months from discharge to those without CO (n = 90), and we assessed the prognostic impact of SUA/eGFR for 12-months CO. RESULTS SUA/eGFR was significantly higher in participants who experienced a CO within 12 months from discharge for AHF, compared to those who did not experience any CO (17.8 (16.6) vs. 13.7 (12.1) mg/dl/ml/min*100, p = 0.008). SUA/eGFR, and not SUA alone, was associated with an increase in the rate of CO (unadjusted HR 1.011, CI 95% 1.004-1.019, p = 0.003). This association lost significance in participants under treatment with xanthine oxidase inhibitors but remained significant after adjustment for multiple confounders. CONCLUSION The SUA/ eGFR ratio provides prognostic value in elderly patients hospitalized for AHF. Future studies may clarify if SUA/eGFR and XOI may represent novel diagnostic and therapeutic approaches for subgroups of patients with AHF.
Collapse
Affiliation(s)
- Federica Piani
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; University of Bologna, Department of Medical and Surgical Sciences, Bologna, Italy.
| | - Samantha Baschino
- University of Bologna, Department of Medical and Surgical Sciences, Bologna, Italy
| | - Davide Agnoletti
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; University of Bologna, Department of Medical and Surgical Sciences, Bologna, Italy
| | - Lucia Calandrini
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Daniela Degli Esposti
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Antonio Di Micoli
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; University of Bologna, Department of Medical and Surgical Sciences, Bologna, Italy
| | - Roberta Falcone
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giulia Fiorini
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; University of Bologna, Department of Medical and Surgical Sciences, Bologna, Italy
| | - Eugenia Ianniello
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Paula Mauloni
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Fulvio Ventura
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Maddalena Veronesi
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; University of Bologna, Department of Medical and Surgical Sciences, Bologna, Italy
| | - Richard J Johnson
- Department of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Claudio Borghi
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; University of Bologna, Department of Medical and Surgical Sciences, Bologna, Italy
| |
Collapse
|
13
|
Kaltsas A, Zikopoulos A, Dimitriadis F, Sheshi D, Politis M, Moustakli E, Symeonidis EN, Chrisofos M, Sofikitis N, Zachariou A. Oxidative Stress and Erectile Dysfunction: Pathophysiology, Impacts, and Potential Treatments. Curr Issues Mol Biol 2024; 46:8807-8834. [PMID: 39194738 DOI: 10.3390/cimb46080521] [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: 07/10/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Erectile dysfunction (ED) is a prevalent condition affecting men's sexual health, with oxidative stress (OS) having recently been identified as a significant contributing causative factor. This narrative review aims to elucidate the role of OS in the pathophysiology of ED, focusing on impact, mechanisms, and potential therapeutic interventions. Key findings indicate that OS disrupts endothelial function and nitric oxide (NO) signaling, crucial for erectile function. Various sources of reactive oxygen species (ROS) and their detrimental effects on penile tissue are discussed, including aging, diabetes mellitus, hypertension, hyperlipidemia, smoking, obesity, alcohol consumption, psychological stress, hyperhomocysteinemia, chronic kidney disease, and sickle cell disease. Major sources of ROS, such as NADPH oxidase, xanthine oxidase, uncoupled endothelial NO synthase (eNOS), and mitochondrial electron transport, are identified. NO is scavenged by these ROS, leading to endothelial dysfunction characterized by reduced NO availability, impaired vasodilation, increased vascular tone, and inflammation. This ultimately results in ED due to decreased blood flow to penile tissue and the inability to achieve or maintain an erection. Furthermore, ROS impact the transmission of nitrergic neurotransmitters by causing the death of nitrergic neurons and reducing the signaling of neuronal NO synthase (nNOS), exacerbating ED. Therapeutic approaches targeting OS, including antioxidants and lifestyle modifications, show promise in ameliorating ED symptoms. The review underscores the need for further research to develop effective treatments, emphasizing the interplay between OS and vascular health in ED. Integrating pharmacological and non-pharmacological strategies could enhance clinical outcomes for ED patients, advocating for OS management in ED treatment protocols to improve patient quality of life.
Collapse
Affiliation(s)
- Aris Kaltsas
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | | | - Fotios Dimitriadis
- Department of Urology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Danja Sheshi
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Magdalena Politis
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Efthalia Moustakli
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Evangelos N Symeonidis
- Department of Urology II, European Interbalkan Medical Center, 55535 Thessaloniki, Greece
| | - Michael Chrisofos
- Third Department of Urology, Attikon University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Nikolaos Sofikitis
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Athanasios Zachariou
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
14
|
Zhang C, Qian H, Cui Y, Li X, Cheng Y, Gao L. Associations between estradiol and hyperuricemia and the mediating effects of TC, TG, and TyG: NHANES 2013-2016. Front Endocrinol (Lausanne) 2024; 15:1422470. [PMID: 39170736 PMCID: PMC11335549 DOI: 10.3389/fendo.2024.1422470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/15/2024] [Indexed: 08/23/2024] Open
Abstract
Objectives To explore the relationship between estradiol (E2) and the incidence of hyperuricemia (HUA) in adult women and to explore whether glucolipid metabolism disorders play a mediating role in mediating this relationship. Methods A total of 2,941 participants aged 20-65 years were included in the National Health and Nutrition Examination Survey (NHANES) 2013-2016. Multivariate logistic regression analysis was performed to evaluate the correlations of E2 with HUA. Multivariate linear regression analysis was performed to evaluate the associations between E2 and triglyceride (TG), total cholesterol (TC), and the triglyceride-glucose index (TyG). The restricted cubic spline (RCS) model was used to further explore the association between E2 and HUA and between TG, TC, and TyG and HUA. Mediation analyses were performed to examine whether TC, TG, and TyG mediated the relationship between E2 and HUA. Results After adjusting for covariates, logistic regression revealed that ln(E2) was significantly associated with HUA in the female subgroup (p = 0.035) and that the incidence of HUA tended to increase with decreasing ln(E2) (p for trend = 0.026). Linear regression showed that E2 was significantly associated with TC (p = 0.032), TG (p = 0.019), and TyG (p = 0.048). The RCS model showed that ln(E2) was linearly correlated with the incidence of HUA (p-overall = 0.0106, p-non-linear = 0.3030). TC and TyG were linearly correlated with HUA (TC: p-overall = 0.0039, p-non-linear = 0.4774; TyG: p-overall = 0.0082, p-non-linear = 0.0663), whereas TG was non-linearly correlated with HUA. Mediation analyses revealed that TC, TG, and TyG significantly mediated the relationship between ln(E2) and HUA (TC, indirect effect: -0.00148, 7.5%, p = 0.008; TG, indirect effect: -0.00062, 3.1%, p = 0.004; TyG, indirect effect: -0.00113, 5.6%, p = 0.016). Conclusion In conclusion, this study demonstrated that compared with women aged 20-45 years, women aged 45-55 years and 55-65 years had lower E2 levels and a greater incidence of HUA. E2 levels and the incidence of HUA were negatively associated in female individuals but not in male individuals. In addition, TC, TG, and TyG, which are markers of glucolipid metabolism, played a mediating role in the association between E2 and HUA.
Collapse
Affiliation(s)
- Chuxin Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hongyang Qian
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yiwei Cui
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaojuan Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuli Cheng
- Qi-Huang Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Lin Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
15
|
Li L, Ye J, Zhao Z, Hu S, Liang H, Ouyang J, Hu Z. Shenfu injection improves isoproterenol-induced heart failure in rats by modulating co-metabolism and regulating the trimethylamine-N-oxide - inflammation axis. Front Pharmacol 2024; 15:1412300. [PMID: 38966553 PMCID: PMC11222397 DOI: 10.3389/fphar.2024.1412300] [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: 04/04/2024] [Accepted: 05/30/2024] [Indexed: 07/06/2024] Open
Abstract
Heart failure (HF) is a chronic condition that progressively worsens and continues to be a major financial burden and public health concern. The "gut-heart" axis provides an innovative perspective and therapeutic strategy for preventing and treating heart failure. Shenfu injection (SFI) is a Traditional Chinese Medicine-based treatment demonstrating potential as a therapeutic strategy for heart failure. However, the precise therapeutic mechanisms of SFI in heart failure are not completely characterized. In this study, HF models were established utilizing subcutaneous multipoint injection of isoproterenol (ISO) at a dosage of 5 mg kg-1·d-1 for 7 days. Serum levels of inflammatory biomarkers were quantified using protein microarrays. Rat feces were analyzed using untargeted metabolomics research and 16S rRNA sequencing. The link between gut microbiota and metabolites was examined using a MetOrigin and Spearman correlation analysis. Our results show that Shenfu injection effectively enhances cardiac function in rats with ISO-induced heart failure by potentially modulating pro-/anti-inflammatory imbalance and reducing serum and urine Trimethylamine-N-oxide (TMAO) levels. Moreover, SFI significantly increases the abundance of Bacteroidota at the phylum level, thereby improving disrupted gut microbiota composition. Additionally, SFI supplementation enriches specific genera known for their capacity to produce short-chain fatty acids. SFI was found to be associated with three key metabolic pathways, as revealed by fecal metabonomics analysis, including the pentose phosphate pathway, pyrimidine metabolism, and purine metabolism. Metabolite tracing analysis revealed that Taurine and hypotaurine metabolism was found to be specific to the microbial community. The biosynthesis of Pyrimidine metabolism, Purine metabolism, beta-alanine metabolism, Naphthalene degradation, Pantothenate, and CoA biosynthesis were identified as co-metabolic pathways between microbes and host. The Spearman correlation analysis was also significantly correlated to differentially expressed metabolites regulated by SFI and the gut microbiota. These results suggest that SFI improves ISO-induced heart failure by modulating co-metabolism and regulating the TMAO-inflammation axis.
Collapse
Affiliation(s)
- Lin Li
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jiahao Ye
- Post-Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhenyu Zhao
- Post-Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Siyuan Hu
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hao Liang
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ji Ouyang
- Post-Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhixi Hu
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
- The Domestic First-class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
16
|
Babijczuk K, Berdzik N, Nowak D, Warżajtis B, Rychlewska U, Starzyk J, Mrówczyńska L, Jasiewicz B. Novel C3-Methylene-Bridged Indole Derivatives with and without Substituents at N1: The Influence of Substituents on Their Hemolytic, Cytoprotective, and Antimicrobial Activity. Int J Mol Sci 2024; 25:5364. [PMID: 38791402 PMCID: PMC11121452 DOI: 10.3390/ijms25105364] [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: 04/15/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Alkaloids are natural compounds useful as scaffolds for discovering new bioactive molecules. This study utilized alkaloid gramine to synthesize two groups of C3-substituted indole derivatives, which were either functionalized at N1 or not. The compounds were characterized by spectroscopic methods. The protective effects of the new compounds against in vitro oxidative hemolysis induced by standard oxidant 2,2'-azobis(2-amidinopropane dihydro chloride (AAPH) on human erythrocytes as a cell model were investigated. Additionally, the compounds were screened for antimicrobial activity. The results indicated that most of the indole derivatives devoid of the N1 substitution exhibited strong cytoprotective properties. The docking studies supported the affinities of selected indole-based ligands as potential antioxidants. Furthermore, the derivatives obtained exhibited potent fungicidal properties. The structures of the eight derivatives possessing indole moiety bridged to the imidazole-, benzimidazole-, thiazole-, benzothiazole-, and 5-methylbenzothiazoline-2-thiones were determined by X-ray diffraction. The C=S bond lengths in the thioamide fragment pointed to the involvement of zwitterionic structures of varying contribution. The predominance of zwitterionic mesomers may explain the lack of cytoprotective properties, while steric effects, which limit multiple the hydrogen-bond acceptor properties of a thione sulfur, seem to be responsible for the high hemolytic activity.
Collapse
Affiliation(s)
- Karolina Babijczuk
- Department of Bioactive Products, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland; (K.B.); (N.B.)
| | - Natalia Berdzik
- Department of Bioactive Products, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland; (K.B.); (N.B.)
| | - Damian Nowak
- Department of Quantum Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland;
| | - Beata Warżajtis
- Department of Crystallography, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland; (B.W.); (U.R.)
| | - Urszula Rychlewska
- Department of Crystallography, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland; (B.W.); (U.R.)
| | - Justyna Starzyk
- Department of Soil Science and Microbiology, Faculty of Agronomy, Horticulture, and Bioengineering, University of Life Science, Szydłowska 50, 60-656 Poznań, Poland;
| | - Lucyna Mrówczyńska
- Department of Cell Biology, Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 6, 61-614 Poznań, Poland;
| | - Beata Jasiewicz
- Department of Bioactive Products, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland; (K.B.); (N.B.)
| |
Collapse
|
17
|
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
|
18
|
Zhou YH, Bai YJ, Zhao XY. Combined exposure to multiple metals on abdominal aortic calcification: results from the NHANES study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:24282-24301. [PMID: 38438641 DOI: 10.1007/s11356-024-32745-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 02/28/2024] [Indexed: 03/06/2024]
Abstract
Exposure to metals increases the risk of many diseases and has become a public health concern. However, few studies have focused on the effect of metal on abdominal aortic calcification (AAC), especially the combined effects of metal mixtures. In this study, we aim to investigate the combined effect of metals on AAC risk and determine the key components in the multiple metals. We tried to investigate the relationship between multiple metal exposure and AAC risk. Fourteen urinary metals were analyzed with five statistical models as follows: generalized linear regression, weighted quantile sum regression (WQS), quantile g-computation (Qgcomp), and Bayesian kernel machine regression (BKMR) models. A total of 838 participants were involved, of whom 241 (28.8%) had AAC. After adjusting for covariates, in multiple metal exposure logistic regression, cadmium (Cd) (OR = 1.364, 95% CI = 1.035-1.797) was positively associated with AAC risk, while cobalt (Co) (OR = 0.631, 95% CI = 0.438-0.908) was negatively associated with AAC risk. A significant positive effect between multiple metal exposure and AAC risk was observed in WQS (OR = 2.090; 95% CI = 1.280-3.420, P < 0.01), Qgcomp (OR = 1.522, 95% CI = 1.012-2.290, P < 0.05), and BKMR models. It was found that the positive association may be driven primarily by Cd, lead (Pb), uranium (U), and tungsten (W). Subgroups analysis showed the association was more significant in participants with BMI ≥ 25 kg/m2, abdominal obesity, drinking, and smoking. Our study shows that exposure to multiple metals increases the risk of AAC in adults aged ≥ 40 years in the USA and that Cd, Pb, U, and W are the main contributors. The association is stronger in participants who are obese, smoker, or drinker.
Collapse
Affiliation(s)
- Yuan-Hang Zhou
- Department of Cardiology, Cardiovascular Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Hereditary Cardiovascular Diseases, Zhengzhou, 450052, China
| | - Yu-Jie Bai
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiao-Yan Zhao
- Department of Cardiology, Cardiovascular Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Henan Key Laboratory of Hereditary Cardiovascular Diseases, Zhengzhou, 450052, China.
| |
Collapse
|
19
|
Griffith JA, King RD, Dunn AC, Lewis SE, Maxwell BA, Nurkiewicz TR, Goldsmith WT, Kelley EE, Bowdridge EC. Maternal nano-titanium dioxide inhalation exposure alters placental cyclooxygenase and oxidant balance in a sexually dimorphic manner. ADVANCES IN REDOX RESEARCH 2024; 10:10.1016/j.arres.2023.100090. [PMID: 38562524 PMCID: PMC10979698 DOI: 10.1016/j.arres.2023.100090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The placenta plays a critical role in nutrient-waste exchange between the maternal and fetal circulation, and thus impacts fetal growth and development. We have previously shown that nano-titanium dioxide (nano-TiO2) inhalation exposure during gestation decreased fetal female pup and placenta mass [1], which persists in the following generation [2]. In utero exposed females, once mated, their offspring's placentas had increased capacity for H2O2 production. Generation of oxidants such as hydrogen peroxide (H2O2), have been shown to impact cyclooxygenase activity, specifically metabolites such as prostacyclin (PGI2) or thromboxane (TXA2). Therefore, we hypothesized that maternal nano-TiO2 inhalation exposure during gestation results in alterations in placental production of prostacyclin and thromboxane mediated by enhanced H2O2 production in a sexually dimorphic manner. Pregnant Sprague-Dawley rats were exposed to nano-TiO2 aerosols or filtered air (sham--control) from gestational day (GD) 10-19. Dams were euthanized on GD 20, and fetal serum and placental tissue were collected based on fetal sex. Fetal placental zones (junctional zone (JZ) and labyrinth zone (LZ)) were assessed for xanthine oxidoreductase (XOR) activity, H2O2, and catalase activity, as well as 6-keto-PGF1α and TXB2 levels. Nano-TiO2 exposed fetal female LZ demonstrated significantly greater XOR activity compared to exposed males. Exposed fetal female LZ also demonstrated significantly diminished catalase activity compared to sham-control females. Exposed fetal female LZ had significantly increased abundance of 6-keto-PGF1α compared to sham-control females and increased TXB2 compared to exposed males. In the aggregate these data indicate that maternal nano-TiO2 inhalation exposure has a greater impact on redox homeostasis and PGI2/TXA2 balance in the fetal female LZ. Future studies need to address if treatment with an XO inhibitor during gestation can prevent diminished fetal female growth during maternal nano-TiO2 inhalation exposure.
Collapse
Affiliation(s)
- Julie A. Griffith
- Department of Physiology, Pharmacology and Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Rachel D. King
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Allison C. Dunn
- Department of Physiology, Pharmacology and Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Sara E. Lewis
- Department of Physiology, Pharmacology and Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Brooke A. Maxwell
- Department of Physiology, Pharmacology and Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Timothy R. Nurkiewicz
- Department of Physiology, Pharmacology and Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - William T. Goldsmith
- Department of Physiology, Pharmacology and Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Eric E. Kelley
- Department of Physiology, Pharmacology and Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Elizabeth C. Bowdridge
- Department of Physiology, Pharmacology and Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
20
|
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
|
21
|
Okesina KB, Odetayo AF, Adeyemi WJ, Ajibare AJ, Okesina AA, Olayaki LA. Naringin from sweet orange peel improves testicular function in high fat diet-induced diabetic rats by modulating xanthine oxidase/uric acid signaling and maintaining redox balance. Lab Anim Res 2024; 40:5. [PMID: 38369526 PMCID: PMC10874537 DOI: 10.1186/s42826-024-00188-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is a metabolic disorder affecting many organs, including the testis. Naringin from orange peel extract (OPE) is a flavanone with fertility-enhancing properties. Hence, this study was designed to establish the effect of naringin on T2DM-induced testicular dysfunction. Thirty male (30) Wistar rats were randomized into five groups control, diabetes, diabetes + naringin, diabetes + OPE, and diabetes + metformin. The administrations were via the oral route and lasted for 28 days. RESULTS Naringin ameliorated T2DM-induced increase in FBS and decrease in serum insulin. It also abrogated T2DM-induced decrease in sperm quality, gonadotropin-releasing hormone, luteinizing hormone, follicle-stimulating hormone, testosterone, estradiol, prolactin, catalase, superoxide dismutase, and total antioxidant capacity. Furthermore, naringin prevented a T2DM-induced increase in malonaldehyde, tumor necrosis factor-alpha, C-reactive protein, xanthine oxidase (XO), and uric acid (UA), it was accompanied by the restoration of normal testicular histoarchitecture. CONCLUSIONS Naringin prevented T2DM-induced testicular dysfunction by modulating XO/UA and restoring redox balance. Also, while the animals treated with OPE exhibited better ameliorative effects than their counterparts treated with naringin, the findings from this study showed that naringin would be a promising supplement for treating T2DM-induced male infertility.
Collapse
Affiliation(s)
- Kazeem Bidemi Okesina
- Department of Medical Physiology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
| | - Adeyemi Fatai Odetayo
- Department of Physiology, Federal University of Health Sciences, Ila Orangun, Nigeria.
| | | | | | - Akeem Ayodeji Okesina
- Department of Clinical Medicine and Community Health, School of Health Sciences, College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
| | | |
Collapse
|
22
|
Gwozdzinski L, Pieniazek A, Gwozdzinski K. Factors Influencing Venous Remodeling in the Development of Varicose Veins of the Lower Limbs. Int J Mol Sci 2024; 25:1560. [PMID: 38338837 PMCID: PMC10855638 DOI: 10.3390/ijms25031560] [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: 12/16/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
One of the early symptoms of chronic venous disease (CVD) is varicose veins (VV) of the lower limbs. There are many etiological environmental factors influencing the development of chronic venous insufficiency (CVI), although genetic factors and family history of the disease play a key role. All these factors induce changes in the hemodynamic in the venous system of the lower limbs leading to blood stasis, hypoxia, inflammation, oxidative stress, proteolytic activity of matrix metalloproteinases (MMPs), changes in microcirculation and, consequently, the remodeling of the venous wall. The aim of this review is to present current knowledge on CVD, including the pathophysiology and mechanisms related to vein wall remodeling. Particular emphasis has been placed on describing the role of inflammation and oxidative stress and the involvement of extracellular hemoglobin as pathogenetic factors of VV. Additionally, active substances used in the treatment of VV were discussed.
Collapse
Affiliation(s)
- Lukasz Gwozdzinski
- Department of Pharmacology and Toxicology, Medical University of Lodz, 90-752 Lodz, Poland;
| | - Anna Pieniazek
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| | - Krzysztof Gwozdzinski
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| |
Collapse
|
23
|
Miller SG, Matias C, Hafen PS, Law AS, Witczak CA, Brault JJ. Uric acid formation is driven by crosstalk between skeletal muscle and other cell types. JCI Insight 2024; 9:e171815. [PMID: 38032735 PMCID: PMC10906236 DOI: 10.1172/jci.insight.171815] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 11/28/2023] [Indexed: 12/02/2023] Open
Abstract
Hyperuricemia is implicated in numerous pathologies, but the mechanisms underlying uric acid production are poorly understood. Using a combination of mouse studies, cell culture studies, and human serum samples, we sought to determine the cellular source of uric acid. In mice, fasting and glucocorticoid treatment increased serum uric acid and uric acid release from ex vivo-incubated skeletal muscle. In vitro, glucocorticoids and the transcription factor FoxO3 increased purine nucleotide degradation and purine release from differentiated muscle cells, which coincided with the transcriptional upregulation of AMP deaminase 3, a rate-limiting enzyme in adenine nucleotide degradation. Heavy isotope tracing during coculture experiments revealed that oxidation of muscle purines to uric acid required their transfer from muscle cells to a cell type that expresses xanthine oxidoreductase, such as endothelial cells. Last, in healthy women, matched for age and body composition, serum uric acid was greater in individuals scoring below average on standard physical function assessments. Together, these studies reveal skeletal muscle purine degradation is an underlying driver of uric acid production, with the final step of uric acid production occurring primarily in a nonmuscle cell type. This suggests that skeletal muscle fiber purine degradation may represent a therapeutic target to reduce serum uric acid and treat numerous pathologies.
Collapse
Affiliation(s)
- Spencer G. Miller
- Indiana Center for Musculoskeletal Health and
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Kinesiology, East Carolina University, Greenville, North Carolina, USA
| | - Catalina Matias
- Indiana Center for Musculoskeletal Health and
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Paul S. Hafen
- Indiana Center for Musculoskeletal Health and
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Andrew S. Law
- Indiana Center for Musculoskeletal Health and
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Carol A. Witczak
- Indiana Center for Musculoskeletal Health and
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jeffrey J. Brault
- Indiana Center for Musculoskeletal Health and
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
24
|
Chenaghlou M, Mahzoon FA, Hamzehzadeh S, Norouzi A, Sahrai H, Mohammadi N, Haghighi NK, Abdollahi M, Sadeghi MT, Banisefid E. Could admission level of uric acid predict total diuretic dose in acute heart failure? BMC Cardiovasc Disord 2024; 24:30. [PMID: 38172681 PMCID: PMC10765671 DOI: 10.1186/s12872-023-03687-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Recent studies have shown that increases in serum UA levels are associated with adverse clinical outcomes in patients with chronic heart failure (CHF); the aim of this study was to determine the relationship between serum uric acid and total diuretic dose received during hospitalization in hospitalized patients with acute exacerbation of heart failure. The main purpose of this study is to determine the role of uric acid as a biomarker that can be a substitute for pro-BNP in clinical evaluation and the need for diuretics in hospitalized patients with acute heart failure. METHODS After approving the plan in the Research Council of the Heart Department and obtaining an ethical code from the Regional Committee on Research Ethics (Human Subjects Studies), the researcher referred to the archives of our center, the case of 100 patients diagnosed with acute heart failure. Cardiac patients were selected, and the information required for the study was collected using a pre-prepared data collection form, and the information was entered into SPSS software after categorization and appropriate analysis and statistical tests were performed on it. Were performed and in all statistical tests the statistical significance level was considered 0.05: RESULTS: 100 patients with acute heart failure were included in this study with a mean age of 63.43 ± 14.78 years. 66% of them were men. The mean dose of furosemide in these patients was 680.92 ± 377.47 mg and the mean serum uric acid level in these patients was 8.55 ± 2.50 mg / dL. In the study of the relationship between the variables, there was a significant relationship between the dose of furosemide received with the serum level of serum uric acid (P = 0.017, r = 0.248 and P = 0.009, r = -0.267, respectively). There is also a significant relationship between serum uric acid level and patient mortality (P = 0.013, r = 0.247). However this relationship lost its significance after multivariate analysis. CONCLUSION There is a significant relationship between serum uric acid level and diuretic use. However, in-hospital mortality is not related to uric acid levels at admission.
Collapse
Affiliation(s)
- Maryam Chenaghlou
- Cardiovascular research center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Abedi Mahzoon
- Student research committee, Tabriz University of Medical Sciences, Tabriz, 5166614756, Iran
| | - Sina Hamzehzadeh
- Student research committee, Tabriz University of Medical Sciences, Tabriz, 5166614756, Iran.
| | - Ali Norouzi
- Student research committee, Tabriz University of Medical Sciences, Tabriz, 5166614756, Iran
| | - Hadi Sahrai
- Student research committee, Tabriz University of Medical Sciences, Tabriz, 5166614756, Iran
| | - Nasibeh Mohammadi
- Faculty of medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Negin Khadem Haghighi
- Student research committee, Tabriz University of Medical Sciences, Tabriz, 5166614756, Iran
| | - Mirsaeed Abdollahi
- Student research committee, Tabriz University of Medical Sciences, Tabriz, 5166614756, Iran
| | | | - Erfan Banisefid
- Student research committee, Tabriz University of Medical Sciences, Tabriz, 5166614756, Iran.
| |
Collapse
|
25
|
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
|
26
|
John SV, Seim GL, Erazo-Flores BJ, Steill J, Freeman J, Votava JA, Arp NL, Qing X, Stewart R, Knoll LJ, Fan J. Macrophages undergo functionally significant reprograming of nucleotide metabolism upon classical activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.27.573447. [PMID: 38234794 PMCID: PMC10793465 DOI: 10.1101/2023.12.27.573447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
During an immune response, macrophages systematically rewire their metabolism in specific ways to support their diversve functions. However, current knowledge of macrophage metabolism is largely concentrated on central carbon metabolism. Using multi-omics analysis, we identified nucleotide metabolism as one of the most significantly rewired pathways upon classical activation. Further isotopic tracing studies revealed several major changes underlying the substantial metabolomic alterations: 1) de novo synthesis of both purines and pyrimidines is shut down at several specific steps; 2) nucleotide degradation activity to nitrogenous bases is increased but complete oxidation of bases is reduced, causing a great accumulation of nucleosides and bases; and 3) cells gradually switch to primarily relying on salvaging the nucleosides and bases for maintaining most nucleotide pools. Mechanistically, the inhibition of purine nucleotide de novo synthesis is mainly caused by nitric oxide (NO)-driven inhibition of the IMP synthesis enzyme ATIC, with NO-independent transcriptional downregulation of purine synthesis genes augmenting the effect. The inhibition of pyrimidine nucleotide de novo synthesis is driven by NO-driven inhibition of CTP synthetase (CTPS) and transcriptional downregulation of thymidylate synthase (TYMS). For the rewiring of degradation, purine nucleoside phosphorylase (PNP) and uridine phosphorylase (UPP) are transcriptionally upregulated, increasing nucleoside degradation activity. However, complete degradation of purine bases by xanthine oxidoreductase (XOR) is inhibited by NO, diverting flux into nucleotide salvage. Inhibiting the activation-induced switch from nucleotide de novo synthesis to salvage by knocking out the purine salvage enzyme hypoxanthine-guanine phosporibosyl transferase (Hprt) significantly alters the expression of genes important for activated macrophage functions, suppresses macrophage migration, and increases pyroptosis. Furthermore, knocking out Hprt or Xor increases proliferation of the intracellular parasite Toxoplasma gondii in macrophages. Together, these studies comprehensively reveal the characteristics, the key regulatory mechanisms, and the functional importance of the dynamic rewiring of nucleotide metabolism in classically activated macrophages.
Collapse
Affiliation(s)
- Steven V John
- Morgridge Institute for Research, Madison, WI
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Gretchen L Seim
- Morgridge Institute for Research, Madison, WI
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Billy J Erazo-Flores
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI
| | - John Steill
- Morgridge Institute for Research, Madison, WI
| | | | | | - Nicholas L Arp
- Morgridge Institute for Research, Madison, WI
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Xin Qing
- Morgridge Institute for Research, Madison, WI
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Ron Stewart
- Morgridge Institute for Research, Madison, WI
| | - Laura J Knoll
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI
| | - Jing Fan
- Morgridge Institute for Research, Madison, WI
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
- Lead contact
| |
Collapse
|
27
|
Lee SCES, Pyo AHA, Koritzinsky M. Longitudinal dynamics of the tumor hypoxia response: From enzyme activity to biological phenotype. SCIENCE ADVANCES 2023; 9:eadj6409. [PMID: 37992163 PMCID: PMC10664991 DOI: 10.1126/sciadv.adj6409] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/23/2023] [Indexed: 11/24/2023]
Abstract
Poor oxygenation (hypoxia) is a common spatially heterogeneous feature of human tumors. Biological responses to tumor hypoxia are orchestrated by the decreased activity of oxygen-dependent enzymes. The affinity of these enzymes for oxygen positions them along a continuum of oxygen sensing that defines their roles in launching reactive and adaptive cellular responses. These responses encompass regulation of all steps in the central dogma, with rapid perturbation of the metabolome and proteome followed by more persistent reprogramming of the transcriptome and epigenome. Core hypoxia response genes and pathways are commonly regulated at multiple inflection points, fine-tuning the dependencies on oxygen concentration and hypoxia duration. Ultimately, shifts in the activity of oxygen-sensing enzymes directly or indirectly endow cells with intrinsic hypoxia tolerance and drive processes that are associated with aggressive phenotypes in cancer including angiogenesis, migration, invasion, immune evasion, epithelial mesenchymal transition, and stemness.
Collapse
Affiliation(s)
- Sandy Che-Eun S. Lee
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Andrea Hye An Pyo
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Marianne Koritzinsky
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
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
|
29
|
Qi J, He P, Yao H, Sun W, Lu P, Qi X, Zhang Z, Jing R, Cui B, Liu D, Ning G. Insulin use and gout risk among patients with type 2 diabetes mellitus: a real-world cohort study in Shanghai, China. Clin Rheumatol 2023; 42:3067-3073. [PMID: 37400692 DOI: 10.1007/s10067-023-06684-8] [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: 11/20/2022] [Revised: 06/07/2023] [Accepted: 06/25/2023] [Indexed: 07/05/2023]
Abstract
OBJECTIVES The effect of insulin use on gout risk remains unknown. This study aimed to investigate the association between insulin use and gout risk among patients with type 2 diabetes mellitus (T2DM). METHODS Based on the Shanghai Link Healthcare Database, patients with newly diagnosed T2DM, with or without insulin exposure, were identified from January 1, 2014 to December 31, 2020, and followed until December 31, 2021. Apart from the original cohort, we also established a 1:2 propensity score-matched cohort. A time-dependent Cox proportional hazards model was used to estimate the hazard ratio (HR) and 95% confidence interval (CI) for gout incidence associated with insulin exposure. RESULTS A total of 414,258 patients with T2DM, including 142,505 insulin users and 271,753 insulin non-users, were enrolled in this study. After a median follow-up of 4.08 years (interquartile range, 2.46-5.90 years), the incidence of gout was significantly higher in insulin users than in insulin non-users (319.35 versus 302.20 cases per 100,000 person-years; HR 1.09, 95% CI 1.03-1.16). The results were robust in propensity score-matched cohort, sensitivity analyses, and stratified analysis of aspirin. In other stratified analyses, the association between insulin use and increased gout risk was found only in patients who were female, or aged 40-69 years, or without hypertension, dyslipidemia, ischemic heart disease, chronic lung disease, kidney disease, or not using diuretic. CONCLUSIONS Insulin use is associated with a significantly increased risk of gout among patients with T2DM. Key Points • The first real-world study to investigate the effect of insulin use on gout risk. • Insulin use is associated with a significantly increased risk of gout among patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Jiying Qi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping He
- Link Healthcare Engineering and Information Department, Shanghai Hospital Development Center, Shanghai, China
| | - Huayan Yao
- Computer Net Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Sun
- Wonders Information Co. Ltd, Shanghai, China
| | - Ping Lu
- Wonders Information Co. Ltd, Shanghai, China
| | - Xiaohui Qi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zizheng Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Renjie Jing
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Cui
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Dongmei Liu
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
30
|
Massimo G, Khambata RS, Chapman T, Birchall K, Raimondi C, Shabbir A, Dyson N, Rathod KS, Borghi C, Ahluwalia A. Natural mutations of human XDH promote the nitrite (NO 2-)-reductase capacity of xanthine oxidoreductase: A novel mechanism to promote redox health? Redox Biol 2023; 67:102864. [PMID: 37713777 PMCID: PMC10511815 DOI: 10.1016/j.redox.2023.102864] [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: 08/06/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/17/2023] Open
Abstract
Several rare genetic variations of human XDH have been shown to alter xanthine oxidoreductase (XOR) activity leading to impaired purine catabolism. However, XOR is a multi-functional enzyme that depending upon the environmental conditions also expresses oxidase activity leading to both O2·- and H2O2 and nitrite (NO2-) reductase activity leading to nitric oxide (·NO). Since these products express important, and often diametrically opposite, biological activity, consideration of the impact of XOR mutations in the context of each aspect of the biochemical activity of the enzyme is needed to determine the potential full impact of these variants. Herein, we show that known naturally occurring hXDH mutations do not have a uniform impact upon the biochemical activity of the enzyme in terms of uric acid (UA), reactive oxygen species (ROS) and nitric oxide ·NO formation. We show that the His1221Arg mutant, in the presence of xanthine, increases UA, O2·- and NO generation compared to the WT, whilst the Ile703Val increases UA and ·NO formation, but not O2·-. We speculate that this change in the balance of activity of the enzyme is likely to endow those carrying these mutations with a harmful or protective influence over health that may explain the current equipoise underlying the perceived importance of XDH mutations. We also show that, in presence of inorganic NO2-, XOR-driven O2·- production is substantially reduced. We suggest that targeting enzyme activity to enhance the NO2--reductase profile in those carrying such mutations may provide novel therapeutic options, particularly in cardiovascular disease.
Collapse
Affiliation(s)
- G Massimo
- William Harvey Research Institute, Barts & the London Faculty of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - R S Khambata
- William Harvey Research Institute, Barts & the London Faculty of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - T Chapman
- LifeArc, Accelerator Building Open Innovation Campus, Stevenage, SG1 2FX, UK
| | - K Birchall
- LifeArc, Accelerator Building Open Innovation Campus, Stevenage, SG1 2FX, UK
| | - C Raimondi
- William Harvey Research Institute, Barts & the London Faculty of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - A Shabbir
- William Harvey Research Institute, Barts & the London Faculty of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Nicki Dyson
- William Harvey Research Institute, Barts & the London Faculty of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - K S Rathod
- William Harvey Research Institute, Barts & the London Faculty of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - C Borghi
- Department of Medical and Surgical Sciences, Faculty of Medicine, University of Bologna, Via Massarenti, N.9, 40138, Italy
| | - A Ahluwalia
- Department of Medical and Surgical Sciences, Faculty of Medicine, University of Bologna, Via Massarenti, N.9, 40138, Italy.
| |
Collapse
|
31
|
Sarakpi T, Mesic A, Speer T. Leukocyte-endothelial interaction in CKD. Clin Kidney J 2023; 16:1845-1860. [PMID: 37915921 PMCID: PMC10616504 DOI: 10.1093/ckj/sfad135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Indexed: 11/03/2023] Open
Abstract
Chronic kidney disease (CKD) represents an independent risk factor for cardiovascular diseases (CVD). Accordingly, CKD patients show a substantial increased risk of cardiovascular mortality. Inflammation represents an important link between CKD and CVD. The interaction between endothelial cells and effector cells of the innate immune system plays a central role in the development and progression of inflammation. Vascular injury causes endothelial dysfunction, leading to augmented oxidative stress, increased expression of leukocyte adhesion molecules and chronic inflammation. CKD induces numerous metabolic changes, creating a uremic milieu resulting in the accumulation of various uremic toxins. These toxins lead to vascular injury, endothelial dysfunction and activation of the innate immune system. Recent studies describe CKD-dependent changes in monocytes that promote endothelial dysfunction and thus CKD progression and CKD-associated CVD. The NLR family pyrin domain containing 3-interleukin-1β-interleukin-6 (NLRP3-IL-1β-IL-6) signaling pathway plays a pivotal role in the development and progression of CVD and CKD alike. Several clinical trials are investigating targeted inhibition of this pathway indicating that anti-inflammatory therapeutic strategies may emerge as novel approaches in patients at high cardiovascular risk and nonresolving inflammation. CKD patients in particular would benefit from targeted anti-inflammatory therapy, since conventional therapeutic regimens have limited efficacy in this population.
Collapse
Affiliation(s)
- Tamim Sarakpi
- Department of Internal Medicine 4 – Nephrology, Goethe University Frankfurt, Frankfurt am Main, Germany
- Else Kröner-Fresenius-Zentrum for Nephrological Research, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Armir Mesic
- Department of Internal Medicine 4 – Nephrology, Goethe University Frankfurt, Frankfurt am Main, Germany
- Else Kröner-Fresenius-Zentrum for Nephrological Research, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Thimoteus Speer
- Department of Internal Medicine 4 – Nephrology, Goethe University Frankfurt, Frankfurt am Main, Germany
- Else Kröner-Fresenius-Zentrum for Nephrological Research, Goethe University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
32
|
Shimizu M, Naito R, Sato A, Ishiwata S, Yatsu S, Shitara J, Matsumoto H, Murata A, Kato T, Suda S, Hiki M, Kuwabara M, Murase T, Nakamura T, Kasai T. Diurnal Variations in Serum Uric Acid, Xanthine, and Xanthine Oxidoreductase Activity in Male Patients with Coronary Artery Disease. Nutrients 2023; 15:4480. [PMID: 37892555 PMCID: PMC10610187 DOI: 10.3390/nu15204480] [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: 09/22/2023] [Revised: 10/01/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Hyperuricemia is influenced by diet and can cause gout. Whether it is a potential risk factor for cardiovascular disease (CVD) remains controversial, and the mechanism is unclear. Similar to CVDs, gout attacks occur more frequently in the morning and at night. A possible reason for this is the diurnal variation in uric acid (UA), However, scientific data regarding this variation in patients with CVD are not available. Thus, we aimed to investigate diurnal variations in serum levels of UA and plasma levels of xanthine, hypoxanthine, and xanthine oxidoreductase (XOR) activity, which were measured at 18:00, 6:00, and 12:00 in male patients with coronary artery disease. Thirty eligible patients participated in the study. UA and xanthine levels significantly increased from 18:00 to 6:00 but significantly decreased from 6:00 to 12:00. By contrast, XOR activity significantly increased both from 18:00 to 6:00 and 6:00 to 12:00. Furthermore, the rates of increase in UA and xanthine levels from night to morning were significantly and positively correlated. In conclusion, UA and xanthine showed similar diurnal variations, whereas XOR activity showed different diurnal variations. The morning UA surge could be due to UA production. The mechanism involved XOR activity, but other factors were also considered.
Collapse
Affiliation(s)
- Megumi Shimizu
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (M.S.); (A.S.); (S.I.); (S.Y.); (J.S.); (H.M.); (A.M.); (T.K.); (S.S.); (M.H.); (T.K.)
- Keiyu Orthopedic Spine and Joint Hospital, Tokyo 120-0015, Japan
| | - Ryo Naito
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (M.S.); (A.S.); (S.I.); (S.Y.); (J.S.); (H.M.); (A.M.); (T.K.); (S.S.); (M.H.); (T.K.)
- Cardiovascular Respiratory Sleep Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Akihiro Sato
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (M.S.); (A.S.); (S.I.); (S.Y.); (J.S.); (H.M.); (A.M.); (T.K.); (S.S.); (M.H.); (T.K.)
| | - Sayaki Ishiwata
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (M.S.); (A.S.); (S.I.); (S.Y.); (J.S.); (H.M.); (A.M.); (T.K.); (S.S.); (M.H.); (T.K.)
| | - Shoichiro Yatsu
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (M.S.); (A.S.); (S.I.); (S.Y.); (J.S.); (H.M.); (A.M.); (T.K.); (S.S.); (M.H.); (T.K.)
| | - Jun Shitara
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (M.S.); (A.S.); (S.I.); (S.Y.); (J.S.); (H.M.); (A.M.); (T.K.); (S.S.); (M.H.); (T.K.)
| | - Hiroki Matsumoto
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (M.S.); (A.S.); (S.I.); (S.Y.); (J.S.); (H.M.); (A.M.); (T.K.); (S.S.); (M.H.); (T.K.)
| | - Azusa Murata
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (M.S.); (A.S.); (S.I.); (S.Y.); (J.S.); (H.M.); (A.M.); (T.K.); (S.S.); (M.H.); (T.K.)
| | - Takao Kato
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (M.S.); (A.S.); (S.I.); (S.Y.); (J.S.); (H.M.); (A.M.); (T.K.); (S.S.); (M.H.); (T.K.)
| | - Shoko Suda
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (M.S.); (A.S.); (S.I.); (S.Y.); (J.S.); (H.M.); (A.M.); (T.K.); (S.S.); (M.H.); (T.K.)
| | - Masaru Hiki
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (M.S.); (A.S.); (S.I.); (S.Y.); (J.S.); (H.M.); (A.M.); (T.K.); (S.S.); (M.H.); (T.K.)
| | - Masanari Kuwabara
- Intensive Care Unit and Department of Cardiology, Toranomon Hospital, Tokyo 105-8470, Japan;
| | - Takayo Murase
- Sanwa Kagaku Kenkyusho Co., Ltd., Inabe 511-0406, Japan; (T.M.); (T.N.)
| | - Takashi Nakamura
- Sanwa Kagaku Kenkyusho Co., Ltd., Inabe 511-0406, Japan; (T.M.); (T.N.)
| | - Takatoshi Kasai
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (M.S.); (A.S.); (S.I.); (S.Y.); (J.S.); (H.M.); (A.M.); (T.K.); (S.S.); (M.H.); (T.K.)
- Cardiovascular Respiratory Sleep Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Sleep and Sleep Disordered Breathing Center, Juntendo University Hospital, Tokyo 113-8431, Japan
| |
Collapse
|
33
|
Shimizu M, Kasai T, Naito R, Sato A, Ishiwata S, Yatsu S, Shitara J, Matsumoto H, Murata A, Kato T, Suda S, Hiki M, Kuwabara M, Murase T, Nakamura T, Daida H. Overnight changes in uric acid, xanthine oxidoreductase and oxidative stress levels and their relationships with sleep-disordered breathing in patients with coronary artery disease. Hypertens Res 2023; 46:2293-2301. [PMID: 37258622 DOI: 10.1038/s41440-023-01331-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 06/02/2023]
Abstract
Serum uric acid (UA) level is associated with the high cumulative incidence or prevalence of coronary artery disease (CAD), and hyperuricemia is considered as an independent risk marker for CAD. Sleep-disordered breathing (SDB) is also associated with an increased risk of CAD. Several studies have shown that SDB is associated with hyperuricemia, but the mechanisms are unclear. We measured serum levels of UA and xanthine oxidoreductase (XOR) activity and urinary levels of 8-hydroxy-2'-deoxyguanosine (8-OHdG), all of which were assessed at 6 p.m. and the following 6 a.m. in males with CAD. In addition, nocturnal pulse oximetry was performed for the night. Overall 32 eligible patients with CAD were enrolled. Serum UA levels significantly increased overnight. (5.32 ± 0.98 mg/dl to 5.46 ± 1.02 mg/dl, p < 0.001) Moreover, XOR activity and urinary 8-OHdG levels significantly increased from 6 p.m. to 6 a.m. Furthermore, 3% Oxygen desaturation index (ODI) was correlated with the overnight changes in XOR activity (r = 0.36, P = 0.047) and urinary 8-OHdG levels (r = 0.41, P = 0.02). In addition, 3%ODI was independently correlated with the changes in XOR activity (correlation coefficient, 0.36; P = 0.047) and 8-OHdG (partial correlation coefficient, 0.63; P = 0.004) in multivariable analyses. SDB severity was associated with the overnight changes in XOR activity and urinary 8-OHdG, suggesting that SDB may be associated with oxidative stress via UA production. This trial is registered at University Hospital Medical Information Network (UMIN), number: UMIN000021624.
Collapse
Affiliation(s)
- Megumi Shimizu
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takatoshi Kasai
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Cardiovascular Respiratory Sleep Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Sleep and Sleep Disordered Breathing Center, Juntendo University Hospital, Tokyo, Japan.
| | - Ryo Naito
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Cardiovascular Respiratory Sleep Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akihiro Sato
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Sayaki Ishiwata
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shoichiro Yatsu
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Jun Shitara
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroki Matsumoto
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Azusa Murata
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takao Kato
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shoko Suda
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masaru Hiki
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masanari Kuwabara
- Intensive Care Unit and Department of Cardiology, Toranomon Hospital, Tokyo, Japan
| | | | | | - Hiroyuki Daida
- Department of Cardiovascular Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
34
|
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
|
35
|
Luo J, Li Y, Chen J, Qiu H, Chen W, Luo X, Chen Y, Tan Y, Li J. Evaluating the role of serum uric acid in the risk stratification and therapeutic response of patients with pulmonary arterial hypertension associated with congenital heart disease (PAH-CHD). Front Pharmacol 2023; 14:1238581. [PMID: 37701027 PMCID: PMC10493272 DOI: 10.3389/fphar.2023.1238581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/09/2023] [Indexed: 09/14/2023] Open
Abstract
Background: Pulmonary arterial hypertension (PAH) is a malignant pulmonary vascular disease that negatively impacts quality of life, exercise capacity, and mortality. This study sought to investigate the relationship between serum uric acid (UA) level and the disease severity and treatment response of patients with PAH and congenital heart disease (PAH-CHD). Methods: This study included 225 CHD patients and 40 healthy subjects. Serum UA was measured in all patients, and UA levels and haemodynamic parameters were re-evaluated in 20 patients who had received PAH-specific drug treatment for at least 7 ± 1 month. Results: Serum UA levels were significantly higher in PAH-CHD patients than in CHD patients with a normal pulmonary artery pressure and normal subjects (347.7 ± 105.7 μmol/L vs. 278.3 ± 84.6 μmol/L; 347.7 ± 105.7 μmol/L vs. 255.7 ± 44.5 μmol/L, p < 0.05). UA levels in the intermediate and high risk groups were significantly higher than those in the low-risk group (365.6 ± 107.8 μmol/L vs. 311.2 ± 82.8 μmol/L; 451.6 ± 117.6 μmol/L vs. 311.2 ± 82.8 μmol/L, p < 0.05). Serum UA levels positively correlated with mean pulmonary arterial pressure, WHO functional class, pulmonary vascular resistance, and NT-proBNP (r = 0.343, 0.357, 0.406, 0.398; p < 0.001), and negatively with mixed venous oxygen saturation (SvO2) and arterial oxygen saturation (SaO2) (r = -0.293, -0.329; p < 0.001). UA significantly decreased from 352.7 ± 97.5 to 294.4 ± 56.8 μmol/L (p = 0.001) after PAH-specific drug treatment for at least 6 months, along with significant decreases in mean pulmonary arterial pressure and pulmonary vascular resistance and increases in cardiac index and mixed SvO2. Conclusion: Serum UA can be used as a practical and economic biomarker for risk stratification and the evaluation of PAH-specific drug treatment effects for patients with PAH-CHD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jiang Li
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
36
|
Handley E, Callanan A. Effects of electrospun fibers containing ascorbic acid on oxidative stress reduction for cardiac tissue engineering. J Appl Polym Sci 2023; 140:e54242. [PMID: 38439767 PMCID: PMC10909520 DOI: 10.1002/app.54242] [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: 01/13/2023] [Revised: 03/17/2023] [Accepted: 05/15/2023] [Indexed: 03/06/2024]
Abstract
Tissue engineering provides promise for regeneration of cardiac tissue following myocardial infarction. However, the harsh microenvironment of the infarct hampers the efficacy of regenerative therapies. Ischemia-reperfusion injury dramatically increases the levels of reactive oxygen species (ROS) within the infarcted area, causing a cascade of further cellular injury. Implantable tissue engineered grafts can target this oxidative stress by delivering pharmaceutical compounds directly into the diseased tissue. Herein, we successfully fabricated electrospun polycaprolactone (PCL) fibers containing varying concentrations of ascorbic acid, a potent antioxidant well known for its ROS-scavenging capabilities. The antioxidant scaffolds displayed significantly improved scavenging of DPPH radicals, superoxide anions and hydroxyl radicals, in a dose dependent manner. Mechanical properties testing indicated that incorporation of ascorbic acid enhanced the strength and Young's modulus of the material, correlating with a moderate but non-significant increase in the crystallinity. Moreover, the scaffolds supported adhesion and maintained survival of human umbilical vein endothelial cells in vitro, indicating good cytocompatibility. These results provide motivation for the use of ascorbic acid-containing fibrous scaffolds to regulate the highly oxidative microenvironment following myocardial infarction.
Collapse
Affiliation(s)
- Ella‐Louise Handley
- Institute for Bioengineering, School of EngineeringUniversity of EdinburghEdinburghUK
| | - Anthony Callanan
- Institute for Bioengineering, School of EngineeringUniversity of EdinburghEdinburghUK
| |
Collapse
|
37
|
Foteva V, Fisher JJ, Qiao Y, Smith R. Does the Micronutrient Molybdenum Have a Role in Gestational Complications and Placental Health? Nutrients 2023; 15:3348. [PMID: 37571285 PMCID: PMC10421405 DOI: 10.3390/nu15153348] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Molybdenum is an essential trace element for human health and survival, with molybdenum-containing enzymes catalysing multiple reactions in the metabolism of purines, aldehydes, and sulfur-containing amino acids. Recommended daily intakes vary globally, with molybdenum primarily sourced through the diet, and supplementation is not common. Although the benefits of molybdenum as an anti-diabetic and antioxidant inducer have been reported in the literature, there are conflicting data on the benefits of molybdenum for chronic diseases. Overexposure and deficiency can result in adverse health outcomes and mortality, although physiological doses remain largely unexplored in relation to human health. The lack of knowledge surrounding molybdenum intake and the role it plays in physiology is compounded during pregnancy. As pregnancy progresses, micronutrient demand increases, and diet is an established factor in programming gestational outcomes and maternal health. This review summarises the current literature concerning varied recommendations on molybdenum intake, the role of molybdenum and molybdoenzymes in physiology, and the contribution these play in gestational outcomes.
Collapse
Affiliation(s)
- Vladimira Foteva
- Mothers and Babies Research Program, Hunter Medical Research Institute, Newcastle, NSW 2305, Australia; (J.J.F.); (R.S.)
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW 2308, Australia
| | - Joshua J. Fisher
- Mothers and Babies Research Program, Hunter Medical Research Institute, Newcastle, NSW 2305, Australia; (J.J.F.); (R.S.)
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW 2308, Australia
| | - Yixue Qiao
- Academy of Pharmacy, Xi’an Jiaotong Liverpool University, Suzhou 215000, China;
| | - Roger Smith
- Mothers and Babies Research Program, Hunter Medical Research Institute, Newcastle, NSW 2305, Australia; (J.J.F.); (R.S.)
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW 2308, Australia
| |
Collapse
|
38
|
Han Y, Cao Y, Han X, Di H, Yin Y, Wu J, Zhang Y, Zeng X. Hyperuricemia and gout increased the risk of long-term mortality in patients with heart failure: insights from the National Health and Nutrition Examination Survey. J Transl Med 2023; 21:463. [PMID: 37438830 DOI: 10.1186/s12967-023-04307-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/25/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND The prevalence of hyperuricemia, gout, and heart failure (HF) is on the rise, and these conditions often share similar risk factors. The present study aimed to evaluate the relationship among hyperuricemia, gout, HF, and all-cause mortality. METHODS The data on nonpregnant participants aged ≥ 20 years with or without hyperuricemia, gout, and HF from the National Health and Nutrition Examination Survey 2001-2018 and 2007-2018 were included in this study. The binary logistic regression, Kaplan-Meier curve, Cox proportional-hazards model, and restricted cubic spline analysis were employed to evaluate the relationship among hyperuricemia, gout, HF, and all-cause mortality. RESULTS Of 204,179,060 and 223,702,171 weighted eligible participants, 40,044,228 (19.6%) and 9,158,600 (4.1%) had hyperuricemia and gout, respectively. Older age, diabetes, stroke, and coronary artery disease were the risk factors for HF among patients with hyperuricemia and gout. The median survival time was 7.00 years and 6.25 years and the 5-year survival rate was 59.9% and 55.9% for patients with HF and hyperuricemia and those with HF and gout, respectively. Patients with hyperuricemia or gout were 2.46 and 2.35 times more likely to have HF and 1.37 and 1.45 times more likely to experience all-cause mortality compared with those who did not exhibit these conditions. The restricted cubic spline showed a nonlinear correlation between uric acid levels and HF and a J-shaped correlation between uric acid levels and all-cause mortality. CONCLUSIONS Ambulatory patients with hyperuricemia or gout were more likely to have HF compared with those without hyperuricemia or gout. Patients with HF with hyperuricemia or gout were more likely to experience all-cause mortality in the long-term follow-up.
Collapse
Affiliation(s)
- Yingdong Han
- Department of family medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases, No. 1 Shuaifuyuan, Beijing, 100730, China
- Division of General Internal Medicine, Department of medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases, No. 1 Shuaifuyuan, Beijing, 100730, China
| | - Yu Cao
- Department of family medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases, No. 1 Shuaifuyuan, Beijing, 100730, China
- Division of General Internal Medicine, Department of medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases, No. 1 Shuaifuyuan, Beijing, 100730, China
| | - Xinxin Han
- Department of family medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases, No. 1 Shuaifuyuan, Beijing, 100730, China
- Division of General Internal Medicine, Department of medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases, No. 1 Shuaifuyuan, Beijing, 100730, China
| | - Hong Di
- Department of family medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases, No. 1 Shuaifuyuan, Beijing, 100730, China
- Division of General Internal Medicine, Department of medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases, No. 1 Shuaifuyuan, Beijing, 100730, China
| | - Yue Yin
- Department of family medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases, No. 1 Shuaifuyuan, Beijing, 100730, China
- Division of General Internal Medicine, Department of medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases, No. 1 Shuaifuyuan, Beijing, 100730, China
| | - Juan Wu
- Department of family medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases, No. 1 Shuaifuyuan, Beijing, 100730, China
- Division of General Internal Medicine, Department of medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases, No. 1 Shuaifuyuan, Beijing, 100730, China
| | - Yun Zhang
- Department of family medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases, No. 1 Shuaifuyuan, Beijing, 100730, China.
- Division of General Internal Medicine, Department of medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases, No. 1 Shuaifuyuan, Beijing, 100730, China.
| | - Xuejun Zeng
- Department of family medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases, No. 1 Shuaifuyuan, Beijing, 100730, China.
- Division of General Internal Medicine, Department of medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, State Key Laboratory of Complex Severe and Rare Diseases, No. 1 Shuaifuyuan, Beijing, 100730, China.
| |
Collapse
|
39
|
Maghsoud Y, Dong C, Cisneros GA. Investigation of the Inhibition Mechanism of Xanthine Oxidoreductase by Oxipurinol: A Computational Study. J Chem Inf Model 2023; 63:4190-4206. [PMID: 37319436 PMCID: PMC10405278 DOI: 10.1021/acs.jcim.3c00624] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Xanthine oxidoreductase (XOR) is an enzyme found in various organisms. It converts hypoxanthine to xanthine and urate, which are crucial steps in purine elimination in humans. Elevated uric acid levels can lead to conditions like gout and hyperuricemia. Therefore, there is significant interest in developing drugs that target XOR for treating these conditions and other diseases. Oxipurinol, an analogue of xanthine, is a well-known inhibitor of XOR. Crystallographic studies have revealed that oxipurinol directly binds to the molybdenum cofactor (MoCo) in XOR. However, the precise details of the inhibition mechanism are still unclear, which would be valuable for designing more effective drugs with similar inhibitory functions. In this study, molecular dynamics and quantum mechanics/molecular mechanics calculations are employed to investigate the inhibition mechanism of XOR by oxipurinol. The study examines the structural and dynamic effects of oxipurinol on the pre-catalytic structure of the metabolite-bound system. Our results provide insights on the reaction mechanism catalyzed by the MoCo center in the active site, which aligns well with experimental findings. Furthermore, the results provide insights into the residues surrounding the active site and propose an alternative mechanism for developing alternative covalent inhibitors.
Collapse
Affiliation(s)
- Yazdan Maghsoud
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Chao Dong
- Department of Chemistry and Physics, The University of Texas Permian Basin, Odessa, Texas 79762, United States
| | - G Andrés Cisneros
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, Texas 75080, United States
- Department of Physics, The University of Texas at Dallas, Richardson, Texas 75080, United States
| |
Collapse
|
40
|
Khurm M, Guo Y, Wu Q, Zhang X, Ghori MU, Rasool MF, Imran I, Saqib F, Wahid M, Guo Z. Conocarpus lancifolius (Combretaceae): Pharmacological Effects, LC-ESI-MS/MS Profiling and In Silico Attributes. Metabolites 2023; 13:794. [PMID: 37512501 PMCID: PMC10385132 DOI: 10.3390/metabo13070794] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/17/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
In folklore medicine, Conocarpus lancifolius is used to treat various illnesses. The main objective of this study was a comprehensive investigation of Conocarpus lancifolius leaf aqueous extract (CLAE) for its antioxidant, cardioprotective, anxiolytic, antidepressant and memory-enhancing capabilities by using different in vitro, in vivo and in silico models. The in vitro experimentation revealed that CLAE consumed an ample amount of total phenolics (67.70 ± 0.15 µg GAE/mg) and flavonoids (47.54 ± 0.45 µg QE/mg) with stronger antiradical effects through DPPH (IC50 = 16.66 ± 0.42 µg/mL), TAC (77.33 ± 0.41 µg AAE/mg) and TRP (79.11 ± 0.67 µg GAE/mg) assays. The extract also displayed suitable acetylcholinesterase (AChE) inhibitory (IC50 = 110.13 ± 1.71 µg/mL) activity through a modified Ellman's method. The toxicology examination presented no mortality or any signs of clinical toxicity in both single-dose and repeated-dose tests. In line with the cardioprotective study, the pretreatment of CLAE was found to be effective in relieving the isoproterenol (ISO)-induced myocardial injury in rats by normalizing the heart weight index, serum cardiac biomarkers, lipid profile and various histopathological variations. In the noise-stress-induced model for behavior attributes, the results demonstrated that CLAE has the tendency to increase the time spent in the central zone and elevated open arms in the open field and elevated plus maze tests (examined for anxiety assessment), reduced periods of immobility in the forced swimming test (for depression) and improved recognition and working memory in the novel object recognition and Morris water maze tests, respectively. Moreover, the LC-ESI-MS/MS profiling predicted 53 phytocompounds in CLAE. The drug-likeness and ADMET analysis exhibited that the majority of the identified compounds have reasonable physicochemical and pharmacokinetic profiles. The co-expression of molecular docking and network analysis indicated that top-ranked CLAE phytoconstituents act efficiently against the key proteins and target multiple signaling pathways to exert its cardiovascular-protectant, anxiolytic, antidepressant and memory-enhancing activity. Hence, this artifact illustrates that the observed biological properties of CLAE elucidate its significance as a sustainable source of bioactive phytochemicals, which appears to be advantageous for pursuing further studies for the development of new therapeutic agents of desired interest.
Collapse
Affiliation(s)
- Muhammad Khurm
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yuting Guo
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Qingqing Wu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xinxin Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Muhammad Umer Ghori
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad 38000, Pakistan
| | - Muhammad Fawad Rasool
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Imran Imran
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Fatima Saqib
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Muqeet Wahid
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Zengjun Guo
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
41
|
Gwozdzinski L, Bernasinska-Slomczewska J, Hikisz P, Wiktorowska-Owczarek A, Kowalczyk E, Pieniazek A. The Effect of Diosmin, Escin, and Bromelain on Human Endothelial Cells Derived from the Umbilical Vein and the Varicose Vein-A Preliminary Study. Biomedicines 2023; 11:1702. [PMID: 37371797 DOI: 10.3390/biomedicines11061702] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 05/29/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
In this study, we investigated the properties of human varicose vein (VV) endothelial cells (HVVEC) in comparison to the human umbilical vein endothelial cells (HUVEC). The cells were treated with three bioactive compounds with proven beneficial effects in the therapy of patients with VV, diosmin, escin, and bromelain. Two concentrations of tested drugs were used (1, 10 mg/mL), which did not affect the viability of either cell type. Escin led to a slight generation of reactive oxygen species in HUVEC cells. We observed a slight release of superoxide in HVVEC cells upon treatment with diosmin and escin. Diosmin and bromelain showed a tendency to release nitric oxide in HUVEC. Using membrane fluorescent probes, we demonstrated a reduced fluidity of HVVEC, which may lead to their increased adhesion, and, consequently, a much more frequent occurrence of venous thrombosis. For the first time, we show the mechanism of action of drugs used in VV therapy on endothelial cells derived from a VV. Studies with HVVEC have shown that tested drugs may lead to a reduction in the adhesive properties of these cells, and thus to a lower risk of thrombosis.
Collapse
Affiliation(s)
- Lukasz Gwozdzinski
- Department of Pharmacology and Toxicology, Medical University of Lodz, 90-752 Lodz, Poland
| | - Joanna Bernasinska-Slomczewska
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
| | - Pawel Hikisz
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
| | | | - Edward Kowalczyk
- Department of Pharmacology and Toxicology, Medical University of Lodz, 90-752 Lodz, Poland
| | - Anna Pieniazek
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
| |
Collapse
|
42
|
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
|
43
|
Miake J, Hisatome I, Tomita K, Isoyama T, Sugihara S, Kuwabara M, Ogino K, Ninomiya H. Impact of Hyper- and Hypo-Uricemia on Kidney Function. Biomedicines 2023; 11:biomedicines11051258. [PMID: 37238929 DOI: 10.3390/biomedicines11051258] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Uric acid (UA) forms monosodium urate (MSU) crystals to exert proinflammatory actions, thus causing gout arthritis, urolithiasis, kidney disease, and cardiovascular disease. UA is also one of the most potent antioxidants that suppresses oxidative stress. Hyper andhypouricemia are caused by genetic mutations or polymorphism. Hyperuricemia increases urinary UA concentration and is frequently associated with urolithiasis, which is augmented by low urinary pH. Renal hypouricemia (RHU) is associated with renal stones by increased level of urinary UA, which correlates with the impaired tubular reabsorption of UA. Hyperuricemia causes gout nephropathy, characterized by renal interstitium and tubular damage because MSU precipitates in the tubules. RHU is also frequently associated with tubular damage with elevated urinary beta2-microglobulin due to increased urinary UA concentration, which is related to impaired tubular UA reabsorption through URAT1. Hyperuricemia could induce renal arteriopathy and reduce renal blood flow, while increasing urinary albumin excretion, which is correlated with plasma xanthine oxidoreductase (XOR) activity. RHU is associated with exercise-induced kidney injury, since low levels of SUA could induce the vasoconstriction of the kidney and the enhanced urinary UA excretion could form intratubular precipitation. A U-shaped association of SUA with organ damage is observed in patients with kidney diseases related to impaired endothelial function. Under hyperuricemia, intracellular UA, MSU crystals, and XOR could reduce NO and activate several proinflammatory signals, impairing endothelial functions. Under hypouricemia, the genetic and pharmacological depletion of UA could impair the NO-dependent and independent endothelial functions, suggesting that RHU and secondary hypouricemia might be a risk factor for the loss of kidney functions. In order to protect kidney functions in hyperuricemic patients, the use of urate lowering agents could be recommended to target SUA below 6 mg/dL. In order to protect the kidney functions in RHU patients, hydration and urinary alkalization may be recommended, and in some cases an XOR inhibitor might be recommended in order to reduce oxidative stress.
Collapse
Affiliation(s)
- Junichiro Miake
- Division of Pharmacology, Department of Pathophysiological and Therapeutic Science, Tottori University Faculty of Medicine, Tottori 683-8503, Japan
| | - Ichiro Hisatome
- Department of Cardiology, Yonago Medical Center, Tottori 683-0006, Japan
| | - Katsuyuki Tomita
- Department of Respiratory Disease, Yonago Medical Center, Tottori 683-0006, Japan
| | - Tadahiro Isoyama
- Department of Urology, Yonago Medical Center, Tottori 683-0006, Japan
| | - Shinobu Sugihara
- Health Service Center, Shimane University, Matsue 690-0823, Japan
| | - Masanari Kuwabara
- Intensive Care Unit and Department of Cardiology, Toranomon Hospital, Tokyo 105-8470, Japan
| | - Kazuhide Ogino
- Department of Cardiology, Tottori Red Cross Hospital, Tottori 680-0017, Japan
| | - Haruaki Ninomiya
- Department of Biological Regulation, Tottori University Faculty of Medicine, Tottori 683-8503, Japan
| |
Collapse
|
44
|
Nunes KZ, Scorza FA, Cavalheiro EA, Vassallo DV. Reduction of vascular reactivity in rat aortas following pilocarpine-induced status epilepticus. Clinics (Sao Paulo) 2023; 78:100195. [PMID: 37099815 PMCID: PMC10149400 DOI: 10.1016/j.clinsp.2023.100195] [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: 10/06/2022] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 04/28/2023] Open
Abstract
OBJECTIVE The authors investigated changes in vascular reactivity in rats following pilocarpine-induced status epilepticus. METHOD Male Wistar rats weighing between 250g and 300g were used. Status epilepticus was induced using 385 mg/kg i.p. pilocarpine. After 40 days the thoracic aorta was dissected and divided into 4 mm rings and the vascular smooth muscle reactivity to phenylephrine was evaluated. RESULTS Epilepsy decreased the contractile responses of the aortic rings to phenylephrine (0.1 nM-300 mM). To investigate if this reduction was induced by increasing NO production with/or hydrogen peroxide L-NAME and Catalase were used. L-NAME (N-nitro-L arginine methyl ester) increased vascular reactivity but the contractile response to phenylephrine increased in the epileptic group. Catalase administration decreased the contractile responses only in the rings of rats with epilepsy. CONCLUSIONS Our findings demonstrated for the first time that epilepsy is capable of causing a reduction of vascular reactivity in rat aortas. These results suggest that vascular reactivity reduction is associated with increased production of Nitric Oxide (NO) as an organic attempt to avoid hypertension produced by excessive sympathetic activation.
Collapse
Affiliation(s)
- Karolini Zuqui Nunes
- Postgraduate Program in Nutrition and Health, Universidade Federal do Espírito Santo, Vitória, ES, Brazil.
| | - Fulvio Alexandre Scorza
- Discipline of Neuroscience, Universidade Federal de São Paulo/Escola Paulista de Medicina, São PauloSP, Brazil
| | - Esper Abrão Cavalheiro
- Discipline of Neuroscience, Universidade Federal de São Paulo/Escola Paulista de Medicina, São PauloSP, Brazil
| | - Dalton Valentim Vassallo
- Graduate Program in Physiological Sciences, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| |
Collapse
|
45
|
Schenkl C, Heyne E, Doenst T, Schulze PC, Nguyen TD. Targeting Mitochondrial Metabolism to Save the Failing Heart. Life (Basel) 2023; 13:life13041027. [PMID: 37109556 PMCID: PMC10143865 DOI: 10.3390/life13041027] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/28/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Despite considerable progress in treating cardiac disorders, the prevalence of heart failure (HF) keeps growing, making it a global medical and economic burden. HF is characterized by profound metabolic remodeling, which mostly occurs in the mitochondria. Although it is well established that the failing heart is energy-deficient, the role of mitochondria in the pathophysiology of HF extends beyond the energetic aspects. Changes in substrate oxidation, tricarboxylic acid cycle and the respiratory chain have emerged as key players in regulating myocardial energy homeostasis, Ca2+ handling, oxidative stress and inflammation. This work aims to highlight metabolic alterations in the mitochondria and their far-reaching effects on the pathophysiology of HF. Based on this knowledge, we will also discuss potential metabolic approaches to improve cardiac function.
Collapse
Affiliation(s)
- Christina Schenkl
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Estelle Heyne
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Torsten Doenst
- Department of Cardiothoracic Surgery, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Paul Christian Schulze
- Department of Medicine I (Cardiology, Angiology, Critical Care Medicine), Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Tien Dung Nguyen
- Department of Medicine I (Cardiology, Angiology, Critical Care Medicine), Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany
| |
Collapse
|
46
|
Abstract
Chronic kidney disease is associated with an increased risk for the development and progression of cardiovascular disorders including hypertension, dyslipidemia, and coronary artery disease. Chronic kidney disease may also affect the myocardium through complex systemic changes, resulting in structural remodeling such as hypertrophy and fibrosis, as well as impairments in both diastolic and systolic function. These cardiac changes in the setting of chronic kidney disease define a specific cardiomyopathic phenotype known as uremic cardiomyopathy. Cardiac function is tightly linked to its metabolism, and research over the past 3 decades has revealed significant metabolic remodeling in the myocardium during the development of heart failure. Because the concept of uremic cardiomyopathy has only been recognized in recent years, there are limited data on metabolism in the uremic heart. Nonetheless, recent findings suggest overlapping mechanisms with heart failure. This work reviews key features of metabolic remodeling in the failing heart in the general population and extends this to patients with chronic kidney disease. The knowledge of similarities and differences in cardiac metabolism between heart failure and uremic cardiomyopathy may help identify new targets for mechanistic and therapeutic research on uremic cardiomyopathy.
Collapse
Affiliation(s)
- T Dung Nguyen
- Department of Internal Medicine I, University Hospital Jena, Jena, Germany
| | | |
Collapse
|
47
|
Capó X, Monserrat-Mesquida M, Quetglas-Llabrés M, Batle JM, Tur JA, Pons A, Sureda A, Tejada S. Hyperbaric Oxygen Therapy Reduces Oxidative Stress and Inflammation, and Increases Growth Factors Favouring the Healing Process of Diabetic Wounds. Int J Mol Sci 2023; 24:ijms24087040. [PMID: 37108205 PMCID: PMC10139175 DOI: 10.3390/ijms24087040] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/09/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Hyperbaric oxygen therapy (HBOT) is the clinical application of oxygen at pressures higher than atmospheric pressure. HBOT has been effectively used to manage diverse clinical pathologies, such as non-healing diabetic ulcers. The aim of the present study was to analyse the effects of HBOT on the plasma oxidative and inflammation biomarkers and growth factors in patients with chronic diabetic wounds. The participants received 20 HBOT sessions (five sessions/week), and blood samples were obtained at sessions 1, 5 and 20, before and 2 h after the HBOT. An additional (control) blood sample was collected 28 days after wound recovery. No significant differences were evident in haematological parameters, whereas the biochemical parameters progressively decreased, which was significant for creatine phosphokinase (CPK) and aspartate aminotransferase (AST). The pro-inflammatory mediators, tumour necrosis factor alpha (TNF-α) and interleukin 1β (IL-1β), progressively decreased throughout the treatments. Biomarkers of oxidative stress--plasma protein levels of catalase, extracellular superoxide dismutase, myeloperoxidase, xanthine oxidase, malondialdehyde (MDA) levels and protein carbonyls--were reduced in accordance with wound healing. Plasma levels of growth factors--platelet-derived growth factor (PDFG), transforming growth factor β (TGF-β) and hypoxia-inducible factor 1-alpha (HIF-1α)-- were increased as a consequence of HBOT and reduced 28 days after complete wound healing, whereas matrix metallopeptidase 9 (MMP9) progressively decreased with the HBOT. In conclusion, HBOT reduced oxidative and pro-inflammatory mediators, and may participate in activating healing, angiogenesis and vascular tone regulation by increasing the release of growth factors.
Collapse
Affiliation(s)
- Xavier Capó
- Research Group in Community Nutrition and Oxidative Stress, University of the Balearic Islands-IUNICS, 07122 Palma, Spain
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
| | - Margalida Monserrat-Mesquida
- Research Group in Community Nutrition and Oxidative Stress, University of the Balearic Islands-IUNICS, 07122 Palma, Spain
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Magdalena Quetglas-Llabrés
- Research Group in Community Nutrition and Oxidative Stress, University of the Balearic Islands-IUNICS, 07122 Palma, Spain
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Juan M Batle
- Research Group in Community Nutrition and Oxidative Stress, University of the Balearic Islands-IUNICS, 07122 Palma, Spain
- MEDISUB Recerca, 07400 Alcúdia, Spain
| | - Josep A Tur
- Research Group in Community Nutrition and Oxidative Stress, University of the Balearic Islands-IUNICS, 07122 Palma, Spain
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Antoni Pons
- Research Group in Community Nutrition and Oxidative Stress, University of the Balearic Islands-IUNICS, 07122 Palma, Spain
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Antoni Sureda
- Research Group in Community Nutrition and Oxidative Stress, University of the Balearic Islands-IUNICS, 07122 Palma, Spain
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Silvia Tejada
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Laboratory of Neurophysiology, Department of Biology, University of the Balearic Islands, 07122 Palma, Spain
| |
Collapse
|
48
|
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
|
49
|
Yu H, Chen X, Guo X, Chen D, Jiang L, Qi Y, Shao J, Tao L, Hang J, Lu G, Chen Y, Li Y. The clinical value of serum xanthine oxidase levels in patients with acute ischemic stroke. Redox Biol 2023; 60:102623. [PMID: 36739755 PMCID: PMC9932569 DOI: 10.1016/j.redox.2023.102623] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/23/2023] [Accepted: 01/31/2023] [Indexed: 02/04/2023] Open
Abstract
Xanthine oxidase (XO), a form of xanthine oxidoreductase, is widely distributed in various human tissues. As a major source for the generation of superoxide radicals, XO is involved in the induction of oxidative stress and inflammation during ischemic and hypoxic tissue injury. Therefore, we designed this study to identify the role of serum XO levels in acute ischemic stroke (AIS) pathogenesis. In this single-center prospective study, 328 consecutive patients with AIS for the first time were included, and 107 age- and sex-matched healthy controls from a community-based stroke screening population were also included. The serum levels of XO and several conventional stroke risk factors were assessed. Multivariate analysis was applied to evaluate the relationship between serum levels of XO and clinical outcomes, and nomogram models were developed to predict the onset, progression and prognosis of AIS. Compared with the healthy control group, the serum level of XO was significantly higher in the AIS group (P < 0.05) and was an independent risk factor for AIS (OR 8.68, 95% CI 4.62-14.33, P < 0.05). Patients with progressive stroke or a poor prognosis had a much higher serum level of XO than patients with stable stroke or a good prognosis (all P < 0.05). In addition, the serum level of XO was an independent risk factor for stroke progression (OR 1.98, 95% CI 1.12-3.50, P = 0.018) and a poor prognosis (OR 2.51, 95% CI 1.47-3.31, P = 0.001). The nomogram models including XO to predict the onset, progression and prognosis of AIS had good prediction and differentiation abilities. The findings of this study show that the serum level of XO on admission was an independent risk factor for AIS and had certain clinical predictive value for stroke progression and prognosis in patients with AIS.
Collapse
Affiliation(s)
- Hailong Yu
- Clinical Medical College of Yangzhou University, Yangzhou, 225001, China,Department of Neuro Intensive Care Unit, Clinical Medical College of Yangzhou University, Yangzhou, 225001, China,Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Xin Chen
- Clinical Medical College of Yangzhou University, Yangzhou, 225001, China,Department of Neuro Intensive Care Unit, Clinical Medical College of Yangzhou University, Yangzhou, 225001, China,Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Xin Guo
- Department of Neurology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, 441100, Xiangyang, China
| | - Danni Chen
- Clinical Medical College of Yangzhou University, Yangzhou, 225001, China,Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Li Jiang
- Clinical Medical College of Yangzhou University, Yangzhou, 225001, China,Department of Geriatrics, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Yajie Qi
- Clinical Medical College of Yangzhou University, Yangzhou, 225001, China,Department of Neuro Intensive Care Unit, Clinical Medical College of Yangzhou University, Yangzhou, 225001, China
| | - Jun Shao
- Clinical Medical College of Yangzhou University, Yangzhou, 225001, China,Department of Cardiac Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Luhang Tao
- Clinical Medical College of Yangzhou University, Yangzhou, 225001, China,Department of Neuro Intensive Care Unit, Clinical Medical College of Yangzhou University, Yangzhou, 225001, China,Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Jing Hang
- Clinical Medical College of Yangzhou University, Yangzhou, 225001, China,Department of Neuro Intensive Care Unit, Clinical Medical College of Yangzhou University, Yangzhou, 225001, China,Department of Neurology, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Guangyu Lu
- School of Public Health, Medical College of Yangzhou University, Yangzhou University, Yangzhou, 225009, China
| | - Yingzhu Chen
- Clinical Medical College of Yangzhou University, Yangzhou, 225001, China; Department of Geriatrics, Northern Jiangsu People's Hospital, Yangzhou, 225001, China.
| | - Yuping Li
- Clinical Medical College of Yangzhou University, Yangzhou, 225001, China; Department of Neuro Intensive Care Unit, Clinical Medical College of Yangzhou University, Yangzhou, 225001, China; Department of Neurosurgery, Clinical Medical College of Yangzhou University, Yangzhou, 225001, China.
| |
Collapse
|
50
|
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
|