1
|
Chen CH, Chen WH, Hsu CP, Lee TS. Atypical antipsychotics impair the lipid-lowering and pleiotropic effects of simvastatin via activation of the ADMA-NOX-ROS pathway. Biomed Pharmacother 2025; 185:117958. [PMID: 40058152 DOI: 10.1016/j.biopha.2025.117958] [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: 12/22/2024] [Revised: 02/20/2025] [Accepted: 03/04/2025] [Indexed: 03/23/2025] Open
Abstract
Patients with schizophrenia receiving atypical antipsychotics have an increased risk of metabolic syndrome; however, the efficacy of statins in mitigating cardiovascular risks in these patients remains unclear. This study examined the effects of typical and atypical antipsychotics on the lipid-lowering efficacy of statins in schizophrenia patients and investigated the underlying mechanisms of simvastatin action in hepatocytes and endothelial cells (ECs). A retrospective analysis revealed that statins were less effective in lowering LDL levels in patients on atypical antipsychotics. In vitro, olanzapine attenuated the beneficial effects of simvastatin in hepatocytes and ECs. Mechanistically, olanzapine downregulated dimethylarginine dimethylaminohydrolase 1 (DDAH1) and/or DDAH2, leading to elevated asymmetric dimethylarginine (ADMA) levels in both cell types. In hepatocytes, olanzapine suppressed low-density lipoprotein receptor (LDLR) expression and reduced LDL binding by activating the NOX-ROS pathway via PPARγ-PCSK9- and LXRα-IDOL-dependent signaling. Inhibition of the NOX-ROS pathway restored LDLR expression, LDL binding, and the lipid-lowering effects of simvastatin. In ECs, olanzapine impaired simvastatin-induced nitric oxide (NO) production and endothelial nitric oxide synthase (eNOS) phosphorylation through NOX-ROS pathway activation. Blocking this pathway reversed eNOS inhibition, restoring the endothelial benefits of simvastatin. Collectively, atypical antipsychotics impair statin efficacy in schizophrenia patients by activating the ADMA-NOX-ROS pathway, which downregulates LDLR in hepatocytes and inhibits eNOS activity in ECs. These findings underscore the need for tailored cardiovascular risk management strategies and identify potential molecular targets to enhance statin effectiveness in patients on atypical antipsychotics.
Collapse
Affiliation(s)
- Chia-Hui Chen
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Hua Chen
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chiao-Po Hsu
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; Faculty of Medicine, School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Tzong-Shyuan Lee
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
2
|
Sedding D, Schmidt TM, Bähre H, Bavendiek U, Casas AI, Chen S, Thao-Vi Dao V, Elbatreek MH, Gutzki F, Hahn A, Kleikers P, Krahn T, Macchiusi C, Martin C, Mucke H, Nogales C, Schmidt BML, Seifert R, Sonnenschein K, Tongers J, Thol J, van der Arend I, van Kuijk SMJ, Wingler K, Wu M, Bauersachs J, McGrath B, Schmidt HHHW. Nutritional L-Citrulline and Tetrahydrobiopterin in Peripheral Artery Disease: A Phase II Randomized Trial (CIPER Study). JACC. ADVANCES 2025; 4:101590. [PMID: 39985883 PMCID: PMC11904498 DOI: 10.1016/j.jacadv.2025.101590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 02/24/2025]
Abstract
BACKGROUND Peripheral artery disease (PAD) is a major public health concern due to its high prevalence, severe impact on individuals' health and quality of life, and substantial economic burden. Pharmacological interventions are still limited with numbers needed-to-treat ranging from 6 (cilostazol) to 50 (aspirin, statins, and vorapaxar). OBJECTIVES This randomized, placebo-controlled, double-blinded crossover interventional trial aims to measure the effect of L-citrulline and tetrahydrobiopterin (H4Bip) on walking distance in patients with PAD, stratified by plasma levels of asymmetric dimethyl L-arginine (ADMA), the endogenous inhibitor of endothelial nitric oxide (NO) synthase. METHODS We measured preinterventional ADMA levels in 51 patients with PAD in Australia and Germany with mean changes in absolute claudication distance (dACD) as the primary outcome upon orally supplementing the L-arginine precursor, L-citrulline (3 g) twice daily for 12 weeks, and, in one arm, additionally H4Bip (0.45 g) once per day for a further 2 weeks. RESULTS Preinterventional ADMA levels were pathological (>0.4 μM) in 34 patients. Supplementation with L-citrulline significantly increased the mean plasma levels of both L-citrulline and L-arginine, from 41.8 ± 2.7 μmol/l to 246.3 ± 67.3 μmol/l (P = 0.004) and from 75.2 ± 4.2 μmol/l to 119.2 ± 6.9 μmol/l (P < 0.0001) respectively, when compared with placebo. dACD in % of control was significantly improved by L-citrulline vs placebo (20.11% ± 4.50% vs 5.73% ± 2.74%, respectively; P = 0.011). Further addition of H4Bip increased the mean percentage dACD to 28.15% ± 6.84% (P = 0.021), but only in patients with preinterventional pathological ADMA levels. CONCLUSIONS L-citrulline and, when ADMA levels are pathological, H4Bip are effective nutritional interventions in patients with PAD warranting further confirmatory trials.
Collapse
Affiliation(s)
- Daniel Sedding
- Klinik für Kardiologie, Universitätsklinikum Halle, Halle, Germany; Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Tim M Schmidt
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Hannover, Germany; Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands
| | - Heike Bähre
- Research Core Unit Metabolomics, Medizinische Hochschule Hannover, Hannover, Germany
| | - Udo Bavendiek
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Ana I Casas
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands
| | - Suzi Chen
- Monash Medical Centre, Monash University, Clayton, Melbourne, Australia
| | - Vu Thao-Vi Dao
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands
| | - Mahmoud H Elbatreek
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Frank Gutzki
- Research Core Unit Metabolomics, Medizinische Hochschule Hannover, Hannover, Germany
| | - Andreas Hahn
- Leibniz Universität Hannover, Institut für Lebensmittelwissenschaft und Humanernährung, Hannover, Germany
| | - Pamela Kleikers
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands
| | - Thomas Krahn
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands
| | - Cecilia Macchiusi
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands; Department of Biostatistics, Sapienza University, Rome, Italy
| | - Catherine Martin
- Monash Medical Centre, Monash University, Clayton, Melbourne, Australia
| | | | - Cristian Nogales
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands
| | - Beate M L Schmidt
- Monash Medical Centre, Monash University, Clayton, Melbourne, Australia
| | - Roland Seifert
- Research Core Unit Metabolomics, Medizinische Hochschule Hannover, Hannover, Germany
| | - Kristina Sonnenschein
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Jörn Tongers
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Jens Thol
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Inés van der Arend
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands
| | - Sander M J van Kuijk
- Clinical Epidemiology and Medical Technology Assessment (KEMTA), Maastricht UMC+, Maastricht, the Netherlands
| | - Kirstin Wingler
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands
| | - Michael Wu
- Monash Medical Centre, Monash University, Clayton, Melbourne, Australia
| | - Johann Bauersachs
- Klinik für Kardiologie und Angiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Barry McGrath
- Monash Medical Centre, Monash University, Clayton, Melbourne, Australia
| | - Harald H H W Schmidt
- Department of Pharmacology and Personalised Medicine, MeHNS, FHML, Maastricht University, Maastricht, the Netherlands; Monash Medical Centre, Monash University, Clayton, Melbourne, Australia.
| |
Collapse
|
3
|
Mu B, Zeng Y, Luo L, Wang K. Oxidative stress-mediated protein sulfenylation in human diseases: Past, present, and future. Redox Biol 2024; 76:103332. [PMID: 39217848 PMCID: PMC11402764 DOI: 10.1016/j.redox.2024.103332] [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: 07/24/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
Reactive Oxygen Species (ROS) refer to a variety of derivatives of molecular oxygen that play crucial roles in regulating a wide range of physiological and pathological processes. Excessive ROS levels can cause oxidative stress, leading to cellular damage and even cell demise. However, moderately elevated levels of ROS can mediate the oxidative post-translational modifications (oxPTMs) of redox-sensitive proteins, thereby affecting protein functions and regulating various cellular signaling pathways. Among the oxPTMs, ROS-induced reversible protein sulfenylation represents the initial form of cysteine oxidation for sensing redox signaling. In this review, we will summarize the discovery, chemical formation, and detection approaches of protein sulfenylation. In addition, we will highlight recent findings for the roles of protein sulfenylation in various diseases, including thrombotic disorders, diabetes, cardiovascular diseases, neurodegenerative diseases, and cancer.
Collapse
Affiliation(s)
- Baoquan Mu
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yan Zeng
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Luo
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, China.
| | - Kui Wang
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
4
|
Vadalà G, Alaimo C, Buccheri G, Di Fazio L, Di Caccamo L, Sucato V, Cipriani M, Galassi AR. Screening and Management of Coronary Artery Disease in Kidney Transplant Candidates. Diagnostics (Basel) 2023; 13:2709. [PMID: 37627968 PMCID: PMC10453389 DOI: 10.3390/diagnostics13162709] [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: 07/13/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Cardiovascular disease (CVD) is a major cause of morbidity and mortality in patients with chronic kidney disease (CKD), especially in end-stage renal disease (ESRD) patients and during the first year after transplantation. For these reasons, and due to the shortage of organs available for transplant, it is of utmost importance to identify patients with a good life expectancy after transplant and minimize the transplant peri-operative risk. Various conditions, such as severe pulmonary diseases, recent myocardial infarction or stroke, and severe aorto-iliac atherosclerosis, need to be ruled out before adding a patient to the transplant waiting list. The effectiveness of systematic coronary artery disease (CAD) treatment before kidney transplant is still debated, and there is no universal screening protocol, not to mention that a nontailored screening could lead to unnecessary invasive procedures and delay or exclude some patients from transplantation. Despite the different clinical guidelines on CAD screening in kidney transplant candidates that exist, up to today, there is no worldwide universal protocol. This review summarizes the key points of cardiovascular risk assessment in renal transplant candidates and faces the role of noninvasive cardiovascular imaging tools and the impact of coronary revascularization versus best medical therapy before kidney transplant on a patient's cardiovascular outcome.
Collapse
Affiliation(s)
- Giuseppe Vadalà
- Division of Cardiology, University Hospital Paolo Giaccone, 90100 Palermo, Italy;
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, 90100 Palermo, Italy; (C.A.); (G.B.); (L.D.F.); (L.D.C.); (A.R.G.)
| | - Chiara Alaimo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, 90100 Palermo, Italy; (C.A.); (G.B.); (L.D.F.); (L.D.C.); (A.R.G.)
| | - Giancarlo Buccheri
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, 90100 Palermo, Italy; (C.A.); (G.B.); (L.D.F.); (L.D.C.); (A.R.G.)
| | - Luca Di Fazio
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, 90100 Palermo, Italy; (C.A.); (G.B.); (L.D.F.); (L.D.C.); (A.R.G.)
| | - Leandro Di Caccamo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, 90100 Palermo, Italy; (C.A.); (G.B.); (L.D.F.); (L.D.C.); (A.R.G.)
| | - Vincenzo Sucato
- Division of Cardiology, University Hospital Paolo Giaccone, 90100 Palermo, Italy;
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, 90100 Palermo, Italy; (C.A.); (G.B.); (L.D.F.); (L.D.C.); (A.R.G.)
| | - Manlio Cipriani
- Institute of Transplant and Highly Specialized Therapies (ISMETT) of Palermo, 90100 Palermo, Italy;
| | - Alfredo Ruggero Galassi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, 90100 Palermo, Italy; (C.A.); (G.B.); (L.D.F.); (L.D.C.); (A.R.G.)
| |
Collapse
|
5
|
Wiedmann MK, Steinsvåg IV, Dinh T, Vigeland MD, Larsson PG, Hjorthaug H, Sheng Y, Mero IL, Selmer KK. Whole-exome sequencing in moyamoya patients of Northern-European origin identifies gene variants involved in Nitric Oxide metabolism: A pilot study. BRAIN & SPINE 2023; 3:101745. [PMID: 37383439 PMCID: PMC10293314 DOI: 10.1016/j.bas.2023.101745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 04/10/2023] [Accepted: 04/21/2023] [Indexed: 06/30/2023]
Abstract
Introduction Moyamoya disease (MMD) is a chronic cerebrovascular steno-occlusive disease of largely unknown etiology. Variants in the RNF213 gene are strongly associated with MMD in East-Asia. In MMD patients of Northern-European origin, no predominant susceptibility variants have been identified so far. Research question Are there specific candidate genes associated with MMD of Northern-European origin, including the known RNF213 gene? Can we establish a hypothesis for MMD phenotype and associated genetic variants identified for further research? Material and methods Adult patients of Northern-European origin, treated surgically for MMD at Oslo University Hospital between October 2018 to January 2019 were asked to participate. WES was performed, with subsequent bioinformatic analysis and variant filtering. The selected candidate genes were either previously reported in MMD or known to be involved in angiogenesis. The variant filtering was based on variant type, location, population frequency, and predicted impact on protein function. Results Analysis of WES data revealed nine variants of interest in eight genes. Five of those encode proteins involved in nitric oxide (NO) metabolism: NOS3, NR4A3, ITGAV, GRB7 and AGXT2. In the AGXT2 gene, a de novo variant was detected, not previously described in MMD. None harboured the p.R4810K missense variant in the RNF213 gene known to be associated with MMD in East-Asian patients. Discussion and conclusion Our findings suggest a role for NO regulation pathways in Northern-European MMD and introduce AGXT2 as a new susceptibility gene. This pilot study warrants replication in larger patient cohorts and further functional investigations.
Collapse
Affiliation(s)
- Markus K.H. Wiedmann
- Department of Neurosurgery, The National Hospital, Oslo University Hospital, Oslo, Norway
| | - Ingunn V. Steinsvåg
- Department of Medical Genetics, Oslo University Hospital and the University of Oslo, Oslo, Norway
| | - Tovy Dinh
- Department of Neurosurgery, The National Hospital, Oslo University Hospital, Oslo, Norway
| | - Magnus D. Vigeland
- Department of Medical Genetics, Oslo University Hospital and the University of Oslo, Oslo, Norway
| | - Pål G. Larsson
- Department of Neurosurgery, The National Hospital, Oslo University Hospital, Oslo, Norway
| | - Hanne Hjorthaug
- Department of Medical Genetics, Oslo University Hospital and the University of Oslo, Oslo, Norway
| | - Ying Sheng
- Department of Medical Genetics, Oslo University Hospital and the University of Oslo, Oslo, Norway
| | - Inger-Lise Mero
- Department of Medical Genetics, Oslo University Hospital and the University of Oslo, Oslo, Norway
| | - Kaja K. Selmer
- Department of Research and Innovation, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
6
|
Huang ST, Yu TM, Chen CH, Cheng YC, Chuang YW, Cheng CH, Liu JS, Hsu CC, Wu MJ. Risk of Major Cardiovascular Disease after Exposure to Contrast Media: A Nationwide Population-Based Cohort Study on Dialysis Patients. Metabolites 2023; 13:metabo13020266. [PMID: 36837885 PMCID: PMC9959650 DOI: 10.3390/metabo13020266] [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: 11/30/2022] [Revised: 01/02/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Contrast associated kidney injury is caused by side effects of iodinated contrast media (ICM), including inflammation. Chronic inflammation among dialysis patient contributes to atherosclerosis, which leads to simultaneous conditions of the kidney, brain, and vasculature. Data to investigate the pathologic effects of ICM on cardiovascular complications in dialysis patients are lacking. Dialysis patients who had been exposed to ICM from computed tomography (ICM-CT) were allocated as the ICM-CT cohort (N = 3751), whereas dialysis patients without ICM exposure were randomly allocated as the non-ICM cohort (N = 17,196). Furthermore, 540 pairs were selected for analyses through propensity score-matching in terms of age, sex, comorbidities, dialysis vintage, and index date. During a median follow-up of 10.3 years, ICM-CT cohort had significantly higher risks in the following, compared with non-ICM cohort: all-cause mortality (adjusted hazard ratio [aHR], 1.36; 95% confidence interval [CI], 1.26-1.47), cardiovascular events (aHR,1.67; 95% CI, 1.39-2.01), acute coronary syndrome (adjusted HR: 2.92; 95% CI, 1.72-4.94), sudden cardiac arrest (aHR, 1.69; 95% CI, 0.90-3.18), heart failure (aHR, 1.71; 95% CI,1.28-2.27), and stroke (aHR, 1.84; 95% CI,1.45-2.35). The proinflammatory ICM is significantly associated with an increased risk of major cardiovascular events in patients on dialysis.
Collapse
Affiliation(s)
- Shih-Ting Huang
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- Graduate Institute of Biomedical Sciences, School of Medicine, China Medical University, Taichung 404333, Taiwan
| | - Tung-Min Yu
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- Graduate Institute of Biomedical Sciences, School of Medicine, China Medical University, Taichung 404333, Taiwan
| | - Chia-Hsin Chen
- Department of Radiology, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Yun-Chung Cheng
- Department of Radiology, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Ya-Wen Chuang
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- Graduate Institute of Biomedical Sciences, School of Medicine, China Medical University, Taichung 404333, Taiwan
| | - Cheng-Hsu Cheng
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- Graduate Institute of Biomedical Sciences, School of Medicine, China Medical University, Taichung 404333, Taiwan
- Department of Life Science, Tunghai University, Taichung 407224, Taiwan
| | - Jia-Sin Liu
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Chih-Cheng Hsu
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli 35053, Taiwan
- Department of Health Services Administration, China Medical University, Taichung 40402, Taiwan
- National Center for Geriatrica and Welfare Research, National Health Research Institiutes, Yunlin 63247, Taiwan
- Department of Family Medicine, Min-Sheng General Hospital, Taoyuan 33044, Taiwan
| | - Ming-Ju Wu
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- RongHsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Correspondence:
| |
Collapse
|
7
|
Yang FM, Shen L, Fan DD, Chen KH, Lee J. DMGV Is a Rheostat of T Cell Survival and a Potential Therapeutic for Inflammatory Diseases and Cancers. Front Immunol 2022; 13:918241. [PMID: 35990633 PMCID: PMC9389583 DOI: 10.3389/fimmu.2022.918241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Activated effector T cells (Teff) and/or compromised regulatory T cells (Treg) underlie many chronic inflammatory diseases. We discovered a novel pathway to regulate survival and expansion of Teff without compromising Treg survival and a potential therapeutic to treat these diseases. We found dimethylguanidino valeric acid (DMGV) as a rheostat for Teff survival: while cell-intrinsic DMGV generated by Alanine-Glyoxylate Aminotransferase 2 (AGXT2) is essential for survival and expansion by inducing mitochondrial ROS and regulation of glycolysis, an excessive (or exogenous) DMGV level inhibits activated Teff survival, thereby the AGXT2-DMGV-ROS axis functioning as a switch to turn on and off Teff expansion. DMGV-induced ROS is essential for glycolysis in Teff, and paradoxically DMGV induces ROS only when glycolysis is active. Mechanistically, DMGV rapidly activates mitochondrial calcium uniporter (MCU), causing a surge in mitochondrial Ca2+ without provoking calcium influx to the cytosol. The mitochondrial Ca2+ surge in turn triggers the mitochondrial Na+/Ca2+ exchanger (NCLX) and the subsequent mitochondrial Na+ import induces ROS by uncoupling the Coenzyme Q cycle in Complex III of the electron transport chain. In preclinical studies, DMGV administration significantly diminished the number of inflammatory T cells, effectively suppressing chronic inflammation in mouse models of colitis and rheumatoid arthritis. DMGV also suppressed expansion of cancer cells in vitro and in a mouse T cell leukemic model by the same mechanism. Our data provide a new pathway regulating T cell survival and a novel mode to treat autoimmune diseases and cancers.
Collapse
Affiliation(s)
- Fengyuan Mandy Yang
- School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, and the State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Liya Shen
- School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, and the State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Dengxia Denise Fan
- School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, and the State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Kuan-Hung Chen
- Department of Orthopedics, The 1st Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Jongdae Lee
- School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, and the State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
8
|
Kurtz T, Pravenec M, DiCarlo S. Mechanism-based strategies to prevent salt sensitivity and salt-induced hypertension. Clin Sci (Lond) 2022; 136:599-620. [PMID: 35452099 PMCID: PMC9069470 DOI: 10.1042/cs20210566] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 12/15/2022]
Abstract
High-salt diets are a major cause of hypertension and cardiovascular (CV) disease. Many governments are interested in using food salt reduction programs to reduce the risk for salt-induced increases in blood pressure and CV events. It is assumed that reducing the salt concentration of processed foods will substantially reduce mean salt intake in the general population. However, contrary to expectations, reducing the sodium density of nearly all foods consumed in England by 21% had little or no effect on salt intake in the general population. This may be due to the fact that in England, as in other countries including the U.S.A., mean salt intake is already close to the lower normal physiologic limit for mean salt intake of free-living populations. Thus, mechanism-based strategies for preventing salt-induced increases in blood pressure that do not solely depend on reducing salt intake merit attention. It is now recognized that the initiation of salt-induced increases in blood pressure often involves a combination of normal increases in sodium balance, blood volume and cardiac output together with abnormal vascular resistance responses to increased salt intake. Therefore, preventing either the normal increases in sodium balance and cardiac output, or the abnormal vascular resistance responses to salt, can prevent salt-induced increases in blood pressure. Suboptimal nutrient intake is a common cause of the hemodynamic disturbances mediating salt-induced hypertension. Accordingly, efforts to identify and correct the nutrient deficiencies that promote salt sensitivity hold promise for decreasing population risk of salt-induced hypertension without requiring reductions in salt intake.
Collapse
Affiliation(s)
- Theodore W. Kurtz
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94017-0134, U.S.A
| | - Michal Pravenec
- Institute of Physiology, Czech Academy of Sciences, Prague 14220, Czech Republic
| | - Stephen E. DiCarlo
- Department of Physiology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, U.S.A
| |
Collapse
|
9
|
Wang D, Wang C, Hao X, Carter G, Carter R, Welch WJ, Wilcox CS. Activation of Nrf2 in Mice Causes Early Microvascular Cyclooxygenase-Dependent Oxidative Stress and Enhanced Contractility. Antioxidants (Basel) 2022; 11:antiox11050845. [PMID: 35624708 PMCID: PMC9137799 DOI: 10.3390/antiox11050845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 12/05/2022] Open
Abstract
Nuclear factor erythroid factor E2-related factor 2 (Nrf2) transcribes antioxidant genes that reduce the blood pressure (BP), yet its activation with tert-butylhydroquinone (tBHQ) in mice infused with angiotensin II (Ang II) increased mean arterial pressure (MAP) over the first 4 days of the infusion. Since tBHQ enhanced cyclooxygenase (COX) 2 expression in vascular smooth muscle cells (VSMCs), we tested the hypothesis that tBHQ administration during an ongoing Ang II infusion causes an early increase in microvascular COX-dependent reactive oxygen species (ROS) and contractility. Mesenteric microarteriolar contractility was assessed on a myograph, and ROS by RatioMaster™. Three days of oral tBHQ administration during the infusion of Ang II increased the mesenteric microarteriolar mRNA for p47phox, the endothelin type A receptor and thromboxane A2 synthase, and increased the excretion of 8-isoprostane F2α and the microarteriolar ROS and contractions to a thromboxane A2 (TxA2) agonist (U-46,619) and endothelin 1 (ET1). These were all prevented in Nrf2 knockout mice. Moreover, the increases in ROS and contractility were prevented in COX1 knockout mice with blockade of COX2 and by blockade of thromboxane prostanoid receptors (TPRs). In conclusion, the activation of Nrf2 over 3 days of Ang II infusion enhances microarteriolar ROS and contractility, which are dependent on COX1, COX2 and TPRs. Therefore, the blockade of these pathways may diminish the early adverse cardiovascular disease events that have been recorded during the initiation of Nrf2 therapy.
Collapse
Affiliation(s)
- Dan Wang
- Division of Nephrology and Hypertension and Hypertension Center, Georgetown University, Washington, DC 20007, USA
| | - Cheng Wang
- Division of Nephrology, Department of Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Xueqin Hao
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang 471023, China
| | - Gabriela Carter
- Division of Nephrology and Hypertension and Hypertension Center, Georgetown University, Washington, DC 20007, USA
| | - Rafaela Carter
- Division of Nephrology and Hypertension and Hypertension Center, Georgetown University, Washington, DC 20007, USA
| | - William J Welch
- Division of Nephrology and Hypertension and Hypertension Center, Georgetown University, Washington, DC 20007, USA
| | - Christopher S Wilcox
- Division of Nephrology and Hypertension and Hypertension Center, Georgetown University, Washington, DC 20007, USA
| |
Collapse
|
10
|
Ebert T, Neytchev O, Witasp A, Kublickiene K, Stenvinkel P, Shiels PG. Inflammation and Oxidative Stress in Chronic Kidney Disease and Dialysis Patients. Antioxid Redox Signal 2021; 35:1426-1448. [PMID: 34006115 DOI: 10.1089/ars.2020.8184] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Significance: Chronic kidney disease (CKD) can be regarded as a burden of lifestyle disease that shares common underpinning features and risk factors with the aging process; it is a complex constituted by several adverse components, including chronic inflammation, oxidative stress, early vascular aging, and cellular senescence. Recent Advances: A systemic approach to tackle CKD, based on mitigating the associated inflammatory, cell stress, and damage processes, has the potential to attenuate the effects of CKD, but it also preempts the development and progression of associated morbidities. In effect, this will enhance health span and compress the period of morbidity. Pharmacological, nutritional, and potentially lifestyle-based interventions are promising therapeutic avenues to achieve such a goal. Critical Issues: In the present review, currents concepts of inflammation and oxidative damage as key patho-mechanisms in CKD are addressed. In particular, potential beneficial but also adverse effects of different systemic interventions in patients with CKD are discussed. Future Directions: Senotherapeutics, the nuclear factor erythroid 2-related factor 2-kelch-like ECH-associated protein 1 (NRF2-KEAP1) signaling pathway, the endocrine klotho axis, inhibitors of the sodium-glucose cotransporter 2 (SGLT2), and live bio-therapeutics have the potential to reduce the burden of CKD and improve quality of life, as well as morbidity and mortality, in this fragile high-risk patient group. Antioxid. Redox Signal. 35, 1426-1448.
Collapse
Affiliation(s)
- Thomas Ebert
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ognian Neytchev
- Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Anna Witasp
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Karolina Kublickiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Paul G Shiels
- Institute of Cancer Sciences, College of Medical, Veterinary & Life Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
11
|
Spence JD. Reducing the Risk of Stroke in Patients with Impaired Renal Function: Nutritional Issues. J Stroke Cerebrovasc Dis 2021; 30:105376. [DOI: 10.1016/j.jstrokecerebrovasdis.2020.105376] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/01/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022] Open
|
12
|
Aliska G, Dhuha A, Rahmatini R, Hamdani R, Afriani N, Tofrizal T, Ali H, Pratiwi N, Nuranisyah V, Mahata LE, Kurnia D. Ramipril Increases Adma Concentration in Acute Myocardial Infarction in Rats Induced by Isoproterenol. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
In experimental animals, the induction of isoproterenol which is a synthetic of catecholamine, can cause acute myocardial infarction where the pathophysiology and morphology are the same as myocardial infarction in humans. Isoproterenol induction will increase oxidative stress, which will damage the enzyme dimethylarginine dimethylaminohydrolase (DDAH), thus causing asymmetric dimethylarginine (ADMA) levels to increase in circulation. Increased levels of ADMA will inhibit the activity of the enzyme nitric oxide synthase, which results in decreased nitric oxide resulting in endothelial damage. This study aims to determine the effect of Ramipril on asymmetric dimethylarginine (ADMA) levels in rats (Rattus norvegicus) Wistar strain with acute myocardial infarction.
Eighteen male Wistar rats (150-250 g) were randomly allocated into three groups: negative control group, positive control, and treatment group. The treatment group was pretreated with Ramipril at dose 3 mg/kg BW orally for seven days. Acute myocardial infarction was induced in positive control groups and treatment groups by subcutaneous injection of isoproterenol (85 mg/kg BW) for two consecutive days. Twenty-four hours after the last administration, rats from all groups were anesthetized and sacrificed for blood sample collection to evaluate the level of Asymmetric Dimethylarginine with Enzym-linked Immunosorbent Assay (ELISA) method. The result showed that ADMA levels were increased in the treatment group after pretreated with Ramipril. This study concluded that pretreatment with Ramipril increased ADMA concentration in acute myocardial infarction rats induced by isoproterenol.
Collapse
|
13
|
Abstract
Dr Irvine Page proposed the Mosaic Theory of Hypertension in the 1940s advocating that hypertension is the result of many factors that interact to raise blood pressure and cause end-organ damage. Over the years, Dr Page modified his paradigm, and new concepts regarding oxidative stress, inflammation, genetics, sodium homeostasis, and the microbiome have arisen that allow further refinements of the Mosaic Theory. A constant feature of this approach to understanding hypertension is that the various nodes are interdependent and that these almost certainly vary between experimental models and between individuals with hypertension. This review discusses these new concepts and provides an introduction to other reviews in this compendium of Circulation Research.
Collapse
Affiliation(s)
- David G. Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center
| | - Thomas M. Coffman
- Cardiovascular and Metabolic Disorders Research Program, Duke-National University of Singapore Medical School
| | | |
Collapse
|
14
|
Low-load resistance training with blood flow restriction prevent renal function decline: The role of the redox balance, angiotensin 1-7 and vasopressin ✰,✰✰. Physiol Behav 2021; 230:113295. [PMID: 33340514 DOI: 10.1016/j.physbeh.2020.113295] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 10/28/2020] [Accepted: 12/12/2020] [Indexed: 02/04/2023]
Abstract
AIMS We sought to investigate the effect of resistance training (RT) and low-load RT with moderate blood flow restriction (RT+BFR) on blood pressure, exercise pressor response, redox balance and vasoactive peptides, body composition and muscle strength in patients with stage two of chronic kidney disease (CKD). METHODS We conducted a 6-month randomized controlled exercise intervention in 90 male and female hypertensive CKD patients (58±9 years with estimated glomerular filtration rate (eGFR; of 66.1 ± 1.2 mL/kg/1.73m2). Participants were randomized to one of three groups (n = 30/group); control group (CTL), RT, and RT+BFR. RT and RT+BFR performed three weekly training sessions using similar periodization for six months (two-month mesocycles), but of different intensities. RESULTS There was similarly effects between RT and RT+BFR in reducing systolic and diastolic blood pressure during daytime and 24hour period (RT: 10.4%; RT+BFR: 10.3% of decrease), fat mass, F2-isoprostanes, asymmetric dimethylarginine (ADMA) and vasopressin (p<0.05 pre-vs post). Also promoted the increase of angiotensin 1-7, nitric oxide (NO), catalase, Trolox equivalent and muscle strength (p<0.05). Both training models attenuated the decline of estimated glomerular filtration rate (p<0.0001 vs CTL). However, only RT+BFR was associated with lower discomfort during exercise (p<0.0001 pre-vs post). Statistical significance was considered with p < 0.05. CONCLUSION These findings suggest low-load RT+BFR as a promising non-pharmacological strategy to control blood pressure, oxidative stress, vasoactive peptides, and consequently, attenuate the decrease of the eGFR.
Collapse
|
15
|
ADMA: A Key Player in the Relationship between Vascular Dysfunction and Inflammation in Atherosclerosis. J Clin Med 2020; 9:jcm9093026. [PMID: 32962225 PMCID: PMC7563400 DOI: 10.3390/jcm9093026] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a chronic cardiovascular disease which increases risk of major cardiovascular events including myocardial infarction and stroke. Elevated plasma concentrations of asymmetric dimethylarginine (ADMA) have long been recognised as a hallmark of cardiovascular disease and are associated with cardiovascular risk factors including hypertension, obesity and hypertriglyceridemia. In this review, we discuss the clinical literature that link ADMA concentrations to increased risk of the development of atherosclerosis. The formation of atherosclerotic lesions relies on the interplay between vascular dysfunction, leading to endothelial activation and the accumulation of inflammatory cells, particularly macrophages, within the vessel wall. Here, we review the mechanisms through which elevated ADMA contributes to endothelial dysfunction, activation and reactive oxygen species (ROS) production; how ADMA may affect vascular smooth muscle phenotype; and finally whether ADMA plays a regulatory role in the inflammatory processes occurring within the vessel wall.
Collapse
|
16
|
New Oxidative Stress Markers Useful in the Diagnosis of Acute Appendicitis in Children: Thiol/Disulfide Homeostasis and the Asymmetric Dimethylarginine Level. Pediatr Emerg Care 2020; 36:362-367. [PMID: 29135899 DOI: 10.1097/pec.0000000000001339] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVES The aim of this study was to evaluate 2 new oxidative stress markers, thiol/disulfide homeostasis status and the asymmetric dimethylarginine (ADMA) level, in children with acute appendicitis (AA) and to evaluate their diagnostic utility. METHODS This case-control study included 45 patients with AA and 35 healthy children. Age, sex, white blood cell count, neutrophil-to-lymphocyte ratio, high-sensitivity C-reactive protein (hs-CRP) level, ultrasonographic findings, thiol/disulfide homeostasis parameters (native and total thiol levels, native thiol/total thiol ratios [antioxidant parameters], and disulfide, disulfide/native thiol, and disulfide/total thiol ratios [oxidant parameters]), and the ADMA level were compared between the 2 groups. RESULTS The native and total thiol levels, and the native thiol/total thiol ratio, were significantly lower, and the disulfide level and disulfide/native thiol and disulfide/total thiol ratios significantly higher, in the AA compared with the control group (all P < 0.001). The ADMA level was significantly higher in a perforated versus nonperforated subgroup of AA patients, but the thiol/disulfide homeostasis parameters did not differ significantly between the two subgroups. In addition, the hs-CRP level and appendiceal wall thickness were higher in the perforated subgroup. The thiol/disulfide antioxidant parameters and ADMA level correlated negatively with the white blood cell count, the neutrophil-to-lymphocyte ratio, and the hs-CRP level, in the AA group, but correlated positively with oxidant parameters. The sensitivity and specificity of the disulfide/native thiol and disulfide/total thiol ratios were high when used to diagnose AA, whereas the sensitivity of the ADMA level was high when used to diagnose perforated appendicitis. CONCLUSIONS Thiol/disulfide homeostasis and the ADMA level, together with certain other parameters, may be useful biomarkers of AA in children.
Collapse
|
17
|
Roumeliotis S, Mallamaci F, Zoccali C. Endothelial Dysfunction in Chronic Kidney Disease, from Biology to Clinical Outcomes: A 2020 Update. J Clin Med 2020; 9:jcm9082359. [PMID: 32718053 PMCID: PMC7465707 DOI: 10.3390/jcm9082359] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023] Open
Abstract
The vascular endothelium is a dynamic, functionally complex organ, modulating multiple biological processes, including vascular tone and permeability, inflammatory responses, thrombosis, and angiogenesis. Endothelial dysfunction is a threat to the integrity of the vascular system, and it is pivotal in the pathogenesis of atherosclerosis and cardiovascular disease. Reduced nitric oxide (NO) bioavailability is a hallmark of chronic kidney disease (CKD), with this disturbance being almost universal in patients who reach the most advanced phase of CKD, end-stage kidney disease (ESKD). Low NO bioavailability in CKD depends on several mechanisms affecting the expression and the activity of endothelial NO synthase (eNOS). Accumulation of endogenous inhibitors of eNOS, inflammation and oxidative stress, advanced glycosylation products (AGEs), bone mineral balance disorders encompassing hyperphosphatemia, high levels of the phosphaturic hormone fibroblast growth factor 23 (FGF23), and low levels of the active form of vitamin D (1,25 vitamin D) and the anti-ageing vasculoprotective factor Klotho all impinge upon NO bioavailability and are critical to endothelial dysfunction in CKD. Wide-ranging multivariate interventions are needed to counter endothelial dysfunction in CKD, an alteration triggering arterial disease and cardiovascular complications in this high-risk population.
Collapse
Affiliation(s)
- Stefanos Roumeliotis
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, School of Medicine, AHEPA Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Francesca Mallamaci
- CNR-IFC (National Research Council of Italy, Centre of Clinical Physiology, Clinical Epidemiology of Renal Diseases and Hypertension Unit, Reggio Cal., c/o Ospedali Riuniti, 89124 Reggio Cal, Italy;
| | - Carmine Zoccali
- CNR-IFC (National Research Council of Italy, Centre of Clinical Physiology, Clinical Epidemiology of Renal Diseases and Hypertension Unit, Reggio Cal., c/o Ospedali Riuniti, 89124 Reggio Cal, Italy;
- Correspondence: ; Tel.: +39-340-73540-62
| |
Collapse
|
18
|
Cozzolino M, Mangano M, Stucchi A, Ciceri P, Conte F, Galassi A. Cardiovascular disease in dialysis patients. Nephrol Dial Transplant 2019; 33:iii28-iii34. [PMID: 30281132 PMCID: PMC6168816 DOI: 10.1093/ndt/gfy174] [Citation(s) in RCA: 349] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular disease (CVD) is a highly common complication and the first cause of death in patients with end-stage renal disease (ESRD) on haemodialysis (HD). In this population, mortality due to CVD is 20 times higher than in the general population and the majority of maintenance HD patients have CVD. This is likely due to ventricular hypertrophy as well as non-traditional risk factors, such as chronic volume overload, anaemia, inflammation, oxidative stress, chronic kidney disease–mineral bone disorder and other aspects of the ‘uraemic milieu’. Better understanding the impact of these numerous factors on CVD would be an important step for prevention and treatment. In this review we focus non-traditional CVD risk factors in HD patients.
Collapse
Affiliation(s)
- Mario Cozzolino
- Department of Health Sciences, Renal Division, University of Milan, San Paolo Hospital, Milan, Italy
| | - Michela Mangano
- Department of Health Sciences, Renal Division, University of Milan, San Paolo Hospital, Milan, Italy
| | - Andrea Stucchi
- Department of Health Sciences, Renal Division, University of Milan, San Paolo Hospital, Milan, Italy
| | - Paola Ciceri
- Renal Research Laboratory Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Ferruccio Conte
- Department of Health Sciences, Renal Division, University of Milan, San Paolo Hospital, Milan, Italy
| | - Andrea Galassi
- Department of Health Sciences, Renal Division, University of Milan, San Paolo Hospital, Milan, Italy
| |
Collapse
|
19
|
Cuevas S, Villar VAM, Jose PA. Genetic polymorphisms associated with reactive oxygen species and blood pressure regulation. THE PHARMACOGENOMICS JOURNAL 2019; 19:315-336. [PMID: 30723314 PMCID: PMC6650341 DOI: 10.1038/s41397-019-0082-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 10/19/2018] [Accepted: 12/21/2018] [Indexed: 02/08/2023]
Abstract
Hypertension is the most prevalent cause of cardiovascular disease and kidney failure, but only about 50% of patients achieve adequate blood pressure control, in part, due to inter-individual genetic variations in the response to antihypertensive medication. Significant strides have been made toward the understanding of the role of reactive oxygen species (ROS) in the regulation of the cardiovascular system. However, the role of ROS in human hypertension is still unclear. Polymorphisms of some genes involved in the regulation of ROS production are associated with hypertension, suggesting their potential influence on blood pressure control and response to antihypertensive medication. This review provides an update on the genes associated with the regulation of ROS production in hypertension and discusses the controversies on the use of antioxidants in the treatment of hypertension, including the antioxidant effects of antihypertensive drugs.
Collapse
Affiliation(s)
- Santiago Cuevas
- Center for Translational Science, Children's National Health System, 111 Michigan Avenue, NW, Washington, DC, 20010, USA.
| | - Van Anthony M Villar
- Department of Medicine, Division of Renal Diseases and Hypertension, The George Washington University School of Medicine and Health Sciences, Walter G. Ross Hall, Suite 738, 2300 I Street, NW, Washington, DC, 20052, USA
| | - Pedro A Jose
- Department of Medicine, Division of Renal Diseases and Hypertension, The George Washington University School of Medicine and Health Sciences, Walter G. Ross Hall, Suite 738, 2300 I Street, NW, Washington, DC, 20052, USA
| |
Collapse
|
20
|
A Pilot Study to Assess Kidney Functions and Toxic Dimethyl-arginines as Risk Biomarkers in Women with Low Vitamin D Levels. J Med Biochem 2019; 38:145-152. [PMID: 30867642 PMCID: PMC6411003 DOI: 10.2478/jomb-2018-0025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/08/2018] [Indexed: 12/26/2022] Open
Abstract
Background Although vitamin D in not a traditional marker for cardiovascular and renal diseases, several studies have proposed a correlation between vitamin D deficiency and these diseases due to the effect of vitamin D on endothelial function. Asymmetric and symmetric dimethyl arginine (ADMA and SDMA, respectively) are endogenous markers of endothelial dysfunction, and are considered as future markers for the assessment of cardiovascular and renal diseases. The present study investigated the association of kidney function tests (urea and creatinine) and dimethylarginine toxins (ADMA and SDMA) in women with vitamin D insufficiency or deficiency. Indeed, sex hormones (estrogen and testosterone) were analyzed in the participants. Methods Women were divided into two groups: premenopausal women (younger than 50 years) and postmenopausal women (older than 50 years). Urea, creatinine, estrogen, testosterone, ADMA, and SDMA levels were analyzed when vitamin D level was deficient or insufficient in the participants. Results The premenopausal women group showed no significant correlations between dimethylarginine toxins and renal failure tests or sex hormones. In the elderly (postmenstrual) women group, only SDMA was significantly correlated with urea and creatinine, while both ADMA and SDMA were not correlated with sex hormones. Conclusions Although ADMA and SDMA are promising candidates of endothelial dysfunction and are increased in menopause and aging, no direct link between ADMA and further progression of renal failure was observed in women with low vitamin D levels. In contrast, a possible direct correlation between SDMA and renal dysfunction was noticed, but only in an age-dependent manner.
Collapse
|
21
|
Patel L, Kilbride HS, Stevens PE, Eaglestone G, Knight S, L Carter J, Delaney MP, Farmer CK, Dalton N, Lamb EJ. Symmetric dimethylarginine is a stronger predictor of mortality risk than asymmetric dimethylarginine among older people with kidney disease. Ann Clin Biochem 2019; 56:367-374. [PMID: 30813746 DOI: 10.1177/0004563218822655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Circulating asymmetric dimethylarginine and symmetric dimethylarginine are increased in patients with kidney disease. Symmetric dimethylarginine is considered a good marker of glomerular filtration rate, while asymmetric dimethylarginine is a marker of cardiovascular risk. However, a link between symmetric dimethylarginine and all-cause mortality has been reported. In the present study, we evaluated both dimethylarginines as risk and glomerular filtration rate markers in a cohort of elderly white individuals, both with and without chronic kidney disease. METHODS Glomerular filtration rate was measured in 394 individuals aged >74 years using an iohexol clearance method. Plasma asymmetric dimethylarginine, symmetric dimethylarginine and iohexol were measured simultaneously using isotope dilution tandem mass spectrometry. RESULTS Plasma asymmetric dimethylarginine concentrations were increased ( P < 0.01) in people with glomerular filtration rate <60 mL/min/1.73 m2 compared with those with glomerular filtration rate ≥60 mL/min/1.73 m2, but did not differ ( P > 0.05) between those with glomerular filtration rate 30-59 mL/min/1.73 m2 and <30 mL/min/1.73 m2. Plasma symmetric dimethylarginine increased consistently across declining glomerular filtration rate categories ( P < 0.0001). Glomerular filtration rate had an independent effect on plasma asymmetric dimethylarginine concentration, while glomerular filtration rate, gender, body mass index and haemoglobin had independent effects on plasma symmetric dimethylarginine concentration. Participants were followed up for a median of 33 months. There were 65 deaths. High plasma asymmetric dimethylarginine ( P = 0.0412) and symmetric dimethylarginine ( P < 0.0001) concentrations were independently associated with reduced survival. CONCLUSIONS Among elderly white individuals with a range of kidney function, symmetric dimethylarginine was a better marker of glomerular filtration rate and a stronger predictor of outcome than asymmetric dimethylarginine. Future studies should further evaluate the role of symmetric dimethylarginine as a marker of outcome and assess its potential value as a marker of glomerular filtration rate.
Collapse
Affiliation(s)
- Liyona Patel
- 1 East Kent Hospitals University NHS Foundation Trust, Canterbury, Kent, UK
| | - Hannah S Kilbride
- 1 East Kent Hospitals University NHS Foundation Trust, Canterbury, Kent, UK
| | - Paul E Stevens
- 1 East Kent Hospitals University NHS Foundation Trust, Canterbury, Kent, UK
| | - Gillian Eaglestone
- 1 East Kent Hospitals University NHS Foundation Trust, Canterbury, Kent, UK
| | - Sarah Knight
- 1 East Kent Hospitals University NHS Foundation Trust, Canterbury, Kent, UK
| | - Joanne L Carter
- 1 East Kent Hospitals University NHS Foundation Trust, Canterbury, Kent, UK
| | - Michael P Delaney
- 1 East Kent Hospitals University NHS Foundation Trust, Canterbury, Kent, UK
| | | | - Neil Dalton
- 2 The Wellchild Laboratory, Evelina London Children's Hospital, London, UK
| | - Edmund J Lamb
- 1 East Kent Hospitals University NHS Foundation Trust, Canterbury, Kent, UK
| |
Collapse
|
22
|
Kurtz TW, DiCarlo SE, Pravenec M, Morris RC. Changing views on the common physiologic abnormality that mediates salt sensitivity and initiation of salt-induced hypertension: Japanese research underpinning the vasodysfunction theory of salt sensitivity. Hypertens Res 2018; 42:6-18. [DOI: 10.1038/s41440-018-0122-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/24/2022]
|
23
|
Dovinová I, Hrabárová E, Jansen E, Kvandová M, Majzúnová M, Berenyiová A, Barančík M. ADMA, homocysteine and redox status improvement affected by 7-nitroindazole in spontaneously hypertensive rats. Biomed Pharmacother 2018; 106:1478-1483. [PMID: 30119222 DOI: 10.1016/j.biopha.2018.07.096] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/18/2018] [Accepted: 07/18/2018] [Indexed: 12/31/2022] Open
Abstract
Inhibition of nitric oxide (NO) production can influence blood pressure regulation and increase hypertension. Asymmetric dimethylarginine, ADMA, an analogue of L-arginine, can inhibit NO synthesis, impair endothelial function, and is a risk marker of cardiovascular diseases. Homocysteine (Hcy) level affects oxidative stress production of reactive oxygen species (ROS) in hypertension and also influences changes in signaling and cell damage. The present study was focused on experimental effects of exogenous NOS inhibitors and their effect on ADMA, an endogenous NOS inhibitor, homocysteine and ROS production measured as reactive oxidative metabolites (ROM). We compared effects of the two potential exogenous NO-inhibitors: NG-nitro L-arginine methyl ester (L-NAME) and 7-nitroindazole (7-NI). Levels of ADMA, Hcy, ROM and total thiols (TTL) were not changed in the L-NAME group. With 7-NI administration, we observed unchanged NOS activity in the left ventricle and a pronounced decrease of ADMA and Hcy levels, accompanied by ROM over-production in plasma. TTL/ROM ratio was more favorable than in the L-NAME group. We observed that 7-NI, an exogenous NOinhibitor, can decrease and improve the levels of ADMA, Hcy, and ROM, and increase TTL/ROM ratio in the plasma of spontaneously hypertensive rats.
Collapse
Affiliation(s)
- Ima Dovinová
- Center of Experimental Medicine - Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Sienkiewiczova 1, 81371 Bratislava, Slovak Republic; Faculty of Health Sciences and Social Work, Trnava University, Univerzitné námestie 1, 918 43 Trnava, Slovak Republic.
| | - Eva Hrabárová
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 9, 84538 Bratislava, Slovak Republic
| | - Eugene Jansen
- Center for Health Protection, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Miroslava Kvandová
- Center of Experimental Medicine - Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Sienkiewiczova 1, 81371 Bratislava, Slovak Republic
| | - Miroslava Majzúnová
- Center of Experimental Medicine - Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Sienkiewiczova 1, 81371 Bratislava, Slovak Republic
| | - Andrea Berenyiová
- Center of Experimental Medicine - Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Sienkiewiczova 1, 81371 Bratislava, Slovak Republic
| | - Miroslav Barančík
- Center of Experimental Medicine - Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 84104 Bratislava, Slovak Republic.
| |
Collapse
|
24
|
Bell T, Araujo M, Luo Z, Tomlinson J, Leiper J, Welch WJ, Wilcox CS. Regulation of fluid reabsorption in rat or mouse proximal renal tubules by asymmetric dimethylarginine and dimethylarginine dimethylaminohydrolase 1. Am J Physiol Renal Physiol 2018; 315:F74-F78. [PMID: 29513072 PMCID: PMC6087787 DOI: 10.1152/ajprenal.00560.2017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 02/15/2018] [Accepted: 03/06/2018] [Indexed: 11/22/2022] Open
Abstract
Nitric oxide prevents hypertension yet enhances proximal tubule Na+ reabsorption. Nitric oxide synthase is inhibited by asymmetric dimethylarginine (ADMA) that is metabolized by dimethylarginine dimethylaminohydrolase (DDAH) whose type 1 isoform is expressed abundantly in the proximal tubule (PT). We hypothesize that ADMA metabolized by DDAH-1 inhibits fluid reabsorbtion (Jv) by the proximal tubule. S2 segments of the PT were microperfused between blocks in vivo to assess Jv in anesthetized rats. Compared with vehicle, microperfusion of ADMA or Nω-nitro-l-arginine methyl ester (l-NAME) in the proximal tubule reduced Jv dose dependently. At 10-4 mol/l both reduced Jv by ~40% (vehicle: 3.2 ± 0.7 vs. ADMA: 2.1 ± 0.5, P < 0.01 vs. l-NAME: 1.9 ± 0.4 nl·min-1·mm-1, P < 0.01; n = 10). Selective inhibition of DDAH-1 in rats with intravenous L-257 (60 mg/kg) given 2 h before and L-257 (10-5 mol/l) perfused in the proximal tubule for 5 min reduced Jv by 32 ± 4% (vehicle: 3.2 ± 0.5 vs. L-257: 2.2 ± 0.5 nl·min-1·mm-1; P < 0.01) and increased plasma ADMA by ≈50% (vehicle: 0.46 ± 0.03 vs. L-257: 0.67 ± 0.03 µmol/l, P < 0.0001) without changing plasma symmetric dimethylarginine. Compared with nontargeted control small-interference RNA, knock down of DDAH-1 in mice by 60% with targeted small-interference RNAs (siRNA) reduced Jv by 29 ± 5% (nontargeted siRNA: 2.8 ± 0.20 vs. DDAH-1 knockdown: 1.9 ± 0.31 nl·min-1·mm-1, P < 0.05). In conclusion, fluid reabsorption in the proximal tubule is reduced by tubular ADMA or by blocking its metabolism by DDAH-1. L-257 is a novel regulator of proximal tubule fluid reabsorption.
Collapse
Affiliation(s)
- Tracy Bell
- Department of Natural Sciences, University of Maryland Eastern Shore, Princess Anne, Maryland
| | - Magali Araujo
- Hypertension Research Center and Division of Nephrology and Hypertension, Georgetown University , Washington, District of Columbia
| | - Zaiming Luo
- Hypertension Research Center and Division of Nephrology and Hypertension, Georgetown University , Washington, District of Columbia
| | - James Tomlinson
- Medical Research Council Clinical Research Center, Royal Postgraduate Medical School and Hammersmith Hospital , London , United Kingdom
| | - James Leiper
- Institute of Cardiovascular and Medical Sciences, University of Glasgow , United Kingdom
| | - William J Welch
- Hypertension Research Center and Division of Nephrology and Hypertension, Georgetown University , Washington, District of Columbia
| | - Christopher S Wilcox
- Hypertension Research Center and Division of Nephrology and Hypertension, Georgetown University , Washington, District of Columbia
| |
Collapse
|
25
|
Wang C, Luo Z, Carter G, Wellstein A, Jose PA, Tomlinson J, Leiper J, Welch WJ, Wilcox CS, Wang D. NRF2 prevents hypertension, increased ADMA, microvascular oxidative stress, and dysfunction in mice with two weeks of ANG II infusion. Am J Physiol Regul Integr Comp Physiol 2017; 314:R399-R406. [PMID: 29167164 DOI: 10.1152/ajpregu.00122.2017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Nuclear factor erythyroid factor 2 (Nrf2) transcribes genes in cultured endothelial cells that reduce reactive oxygen species (ROS) and generate nitric oxide (NO) or metabolize asymmetric dimethylarginine (ADMA), which inhibits NO synthase (NOS). Therefore, we undertook a functional study to test the hypothesis that activation of Nrf2 by tert-butylhydroquinone (tBHQ) preserves microvascular endothelial function during oxidative stress. Wild-type CB57BL/6 (wt), Nrf2 wt (+/+), or knockout (-/-) mice received vehicle (Veh) or tBHQ (0.1%; activator of Nrf2) during 14-day infusions of ANG II (to induce oxidative stress) or sham. MAP was recorded by telemetry. Mesenteric resistance arterioles were studied on isometric myographs and vascular NO and ROS by fluorescence microscopy. ANG II increased the mean arterial pressure (112 ± 5 vs. 145 ± 5 mmHg; P < 0.01) and excretion of 8-isoprostane F2α (2.8 ± 0.3 vs. 3.8 ± 0.3 ng/mg creatinine; P < 0.05) at 12-14 days. However, 12 days of ANG II reduced endothelium-derived relaxation (27 ± 5 vs. 17 ± 3%; P < 0.01) and NO (0.38 ± 0.07 vs. 0.18 ± 0.03 units; P < 0.01) but increased microvascular remodeling, endothelium-derived contractions (7.5 ± 0.5 vs. 13.0 ± 1.7%; P < 0.01), superoxide (0.09 ± 0.03 vs. 0.29 ± 0.08 units; P < 0.05), and contractions to U-46,619 (87 ± 6 vs. 118 ± 3%; P < 0.05), and endothelin-1(89 ± 4 vs. 123 ± 12%; P < 0.05). tBHQ prevented all of these effects of ANG II at 12-14 days in Nrf2+/+ mice but not in Nrf2-/- mice. In conclusion, tBHQ activates Nrf2 to prevent microvascular endothelial dysfunction, remodeling, and contractility, and moderate ADMA and hypertension at 12-14 days of ANG II infusion, thereby preserving endothelial function and preventing hypertension.
Collapse
Affiliation(s)
- Cheng Wang
- Hypertension Center and Division of Nephrology and Hypertension, Georgetown University , Washington, D.C.,Division of Nephrology, Department of Medicine, 5th Hospital of Sun Yat-Sen University , Zhuhai, Guangdong , China
| | - Zaiming Luo
- Hypertension Center and Division of Nephrology and Hypertension, Georgetown University , Washington, D.C
| | - Gabriella Carter
- Hypertension Center and Division of Nephrology and Hypertension, Georgetown University , Washington, D.C
| | - Anton Wellstein
- Lombardi Cancer Center, Georgetown University , Washington, D.C
| | - Pedro A Jose
- Division of Nephrology, George Washington University School of Medicine and Health Sciences , Washington, D.C
| | - James Tomlinson
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College , London , United Kingdom
| | - James Leiper
- Institute of Cardiovascular and Medical Sciences , University of Glasgow , Glasgow United Kingdom
| | - William J Welch
- Hypertension Center and Division of Nephrology and Hypertension, Georgetown University , Washington, D.C
| | - Christopher S Wilcox
- Hypertension Center and Division of Nephrology and Hypertension, Georgetown University , Washington, D.C
| | - Dan Wang
- Hypertension Center and Division of Nephrology and Hypertension, Georgetown University , Washington, D.C
| |
Collapse
|
26
|
Decreased diabetes risk over 9 year after 18-month oral L-arginine treatment in middle-aged subjects with impaired glucose tolerance and metabolic syndrome (extension evaluation of L-arginine study). Eur J Nutr 2017; 57:2805-2817. [PMID: 29052766 PMCID: PMC6267392 DOI: 10.1007/s00394-017-1548-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/25/2017] [Indexed: 01/06/2023]
Abstract
Purpose This study aimed to determine whether l-arginine supplementation lasting for 18 months maintained long-lasting effects on diabetes incidence, insulin secretion and sensitivity, oxidative stress, and endothelial function during 108 months among subjects at high risk of developing type 2 diabetes. Methods One hundred and forty-four middle-aged subjects with impaired glucose tolerance and metabolic syndrome were randomized in 2006 to an l-arginine supplementation (6.4 g orally/day) or placebo therapy lasting 18 months. This period was followed by a 90-month follow-up. The primary outcome was a diagnosis of diabetes during the 108 month study period. Secondary outcomes included changes in insulin secretion (proinsulin/c-peptide ratio), insulin sensitivity (IGI/HOMA-IR), oxidative stress (AOPPs), and vascular function. After the 18 month participation, subjects that were still free of diabetes and willing to continue their participation (104 subjects) were further followed until diabetes diagnosis, with a time span of about 9 years from baseline. Results Although results derived from the 18 month of the intervention study demonstrated no differences in the probability of becoming diabetics, at the end of the study, the cumulative incidence of diabetes was of 40.6% in the l-arginine group and of 57.4% in the placebo group. The adjusted HR for diabetes (l-arginine vs. placebo) was 0.66; 95% CI 0.48, 0.91; p < 0.02). Proinsulin/c-peptide ratio (p < 0.001), IGI/HOMA-IR (p < 0.01), and AOPP (p < 0.05) levels were ameliorated in l-arginine compared to placebo. Conclusions These results may suggest that the administration of l-arginine could delay the development of T2DM for a long period. This effect could be mediated, in some extent, by l-arginine-induced reduction in oxidative stress.
Collapse
|
27
|
Vanhoutte PM, Zhao Y, Xu A, Leung SWS. Thirty Years of Saying NO: Sources, Fate, Actions, and Misfortunes of the Endothelium-Derived Vasodilator Mediator. Circ Res 2017; 119:375-96. [PMID: 27390338 DOI: 10.1161/circresaha.116.306531] [Citation(s) in RCA: 289] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 06/02/2016] [Indexed: 12/16/2022]
Abstract
Endothelial cells control vascular tone by releasing nitric oxide (NO) produced by endothelial NO synthase. The activity of endothelial NO synthase is modulated by the calcium concentration and by post-translational modifications (eg, phosphorylation). When NO reaches vascular smooth muscle, soluble guanylyl cyclase is its primary target producing cGMP. NO production is stimulated by circulating substances (eg, catecholamines), platelet products (eg, serotonin), autacoids formed in (eg, bradykinin) or near (eg, adiponectin) the vascular wall and physical factors (eg, shear stress). NO dysfunction can be caused, alone or in combination, by abnormal coupling of endothelial cell membrane receptors, insufficient supply of substrate (l-arginine) or cofactors (tetrahydrobiopterin), endogenous inhibitors (asymmetrical dimethyl arginine), reduced expression/presence/dimerization of endothelial NO synthase, inhibition of its enzymatic activity, accelerated disposition of NO by reactive oxygen species and abnormal responses (eg, biased soluble guanylyl cyclase activity producing cyclic inosine monophosphate) of the vascular smooth muscle. Major culprits causing endothelial dysfunction, irrespective of the underlying pathological process (aging, obesity, diabetes mellitus, and hypertension), include stimulation of mineralocorticoid receptors, activation of endothelial Rho-kinase, augmented presence of asymmetrical dimethyl arginine, and exaggerated oxidative stress. Genetic and pharmacological interventions improve dysfunctional NO-mediated vasodilatations if protecting the supply of substrate and cofactors for endothelial NO synthase, preserving the presence and activity of the enzyme and reducing reactive oxygen species generation. Common achievers of such improvement include maintained levels of estrogens and increased production of adiponectin and induction of silent mating-type information regulation 2 homologue 1. Obviously, endothelium-dependent relaxations are not the only beneficial action of NO in the vascular wall. Thus, reduced NO-mediated responses precede and initiate the atherosclerotic process.
Collapse
Affiliation(s)
- Paul M Vanhoutte
- From the State Key Laboratory of Pharmaceutical Biotechnology (P.M.V., Y.Z., A.X., S.W.S.L.), Department of Pharmacology and Pharmacy (P.M.V., Y.Z., A.X., S.W.S.L.), and Department of Medicine (A.X.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Yingzi Zhao
- From the State Key Laboratory of Pharmaceutical Biotechnology (P.M.V., Y.Z., A.X., S.W.S.L.), Department of Pharmacology and Pharmacy (P.M.V., Y.Z., A.X., S.W.S.L.), and Department of Medicine (A.X.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- From the State Key Laboratory of Pharmaceutical Biotechnology (P.M.V., Y.Z., A.X., S.W.S.L.), Department of Pharmacology and Pharmacy (P.M.V., Y.Z., A.X., S.W.S.L.), and Department of Medicine (A.X.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Susan W S Leung
- From the State Key Laboratory of Pharmaceutical Biotechnology (P.M.V., Y.Z., A.X., S.W.S.L.), Department of Pharmacology and Pharmacy (P.M.V., Y.Z., A.X., S.W.S.L.), and Department of Medicine (A.X.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
28
|
Sangodele JO, Olaleye MT, Monsees TK, Akinmoladun AC. The para isomer of dinitrobenzene disrupts redox homeostasis in liver and kidney of male wistar rats. Biochem Biophys Rep 2017; 10:297-302. [PMID: 28955757 PMCID: PMC5614678 DOI: 10.1016/j.bbrep.2017.04.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 03/24/2017] [Accepted: 04/26/2017] [Indexed: 02/07/2023] Open
Abstract
Background Para-Dinitrobenzene (p-DNB) is one of the isomers of dinitrobenzene which have been detected as environmental toxicants. Skin irritation and organ toxicities are likely for industrial workers exposed to p-DNB. This study evaluated the effect of sub-chronic exposure of rats to p-DNB on cellular redox balance, hepatic and renal integrity. Methods Forty eight male Wistar rats weighing 160–180 g were administered 50, 75, 1000 and 2000 mg/kg b.wt (body weight) of p-DNB or an equivalent volume of vehicle (control) orally and topically for 14 days. After the period of treatment, the activities of kidney and liver catalase (CAT), alkaline phosphatase (ALP) and superoxide dismutase (SOD) as well as extent of renal and hepatic lipid peroxidation (LPO) were determined. Serum ALP activity and plasma urea concentration were also evaluated. Results Compared with control animals, p-DNB -administered rats showed decrease in the body and relative kidney and liver weights as well as increased renal and hepatic hydrogen peroxide and lipid peroxidation levels accompanied by decreased superoxide dismutase and catalase activities. However, p-DNB caused a significant increase in plasma urea concentration and serum, liver and kidney ALP activities relative to control. In addition, p-DNB caused periportal infiltration, severe macro vesicular steatosis and hepatic necrosis in the liver. Conclusions Our findings show that sub-chronic oral and sub-dermal administration of p-DNB may produce hepato-nephrotoxicity through oxidative stress. Activities of kidney and liver catalase and superoxide dismutase were decreased by p-DNB. p-DNB increased serum, liver and kidney activity of alkaline phosphatase. Plasma urea concentration was increased by p-DNB. Lipid peroxidation and H2O2 level were increased by p-DNB. p-DNB caused histopathological changes in liver and kidney tissues.
Collapse
Key Words
- ALP, alanine phosphatase
- CAT, Catalase
- Environmental toxicants
- GSH, glutathione
- GST, glutathione –s –transferase, GPX, glutathione reductase, NIH, national institute of health
- H&E, hamatoxilin eosin
- Kidney
- LPO, lipid peroxidation
- Liver
- MDA, malodialdehyde
- OECD, Organisation for economic co-operation and Development
- Oxidative stress
- PHS, public health service
- SOD, Superoxide dismutase
- SPSS, Statistical Pucteage for Social Sciences
- Sub-dermal
- TBA, thiobarbituric acid
- TNB, trinitrobenzene
- o-DNB, ortho-dinitrobenzene, m-DNB, meta-dinitrobenzene
- p-DNB, para-dinitrobenzene
- p‐DNB
Collapse
Affiliation(s)
- Janet Olayemi Sangodele
- Phytomedicine, Biochemical Pharmacology and Toxicology Laboratories, Department of Biochemistry, School of Sciences, PMB 704, The Federal University of Technology, Zip code: 340001, Akure, Nigeria.,Department of Medical Biosciences, 4th floor, Life Science Building, Faculty of Natural Science, University of the Western Cape, Bellville, Cape Town, South Africa
| | - Mary Tolulope Olaleye
- Phytomedicine, Biochemical Pharmacology and Toxicology Laboratories, Department of Biochemistry, School of Sciences, PMB 704, The Federal University of Technology, Zip code: 340001, Akure, Nigeria
| | - Thomas K Monsees
- Department of Medical Biosciences, 4th floor, Life Science Building, Faculty of Natural Science, University of the Western Cape, Bellville, Cape Town, South Africa
| | - Afolabi Clement Akinmoladun
- Phytomedicine, Biochemical Pharmacology and Toxicology Laboratories, Department of Biochemistry, School of Sciences, PMB 704, The Federal University of Technology, Zip code: 340001, Akure, Nigeria
| |
Collapse
|
29
|
Interplay between Oxidative Stress and Nutrient Sensing Signaling in the Developmental Origins of Cardiovascular Disease. Int J Mol Sci 2017; 18:ijms18040841. [PMID: 28420139 PMCID: PMC5412425 DOI: 10.3390/ijms18040841] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 04/04/2017] [Accepted: 04/13/2017] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD) presents a global health burden, despite recent advances in management. CVD can originate from early life by so-called “developmental origins of health and disease” (DOHaD). Epidemiological and experimental evidence supports that early-life insults can induce programming of later CVD. Underlying the DOHaD concept, early intervention may offset programming process to prevent the development of CVD, namely reprogramming. Oxidative stress and nutrient sensing signals have been considered to be major mechanisms of cardiovascular programming, while the interplay between these two mechanisms have not been examined in detail. This review summarizes current evidence that supports the link between oxidative stress and nutrient sensing signaling to cardiovascular programming, with an emphasis on the l-arginine–asymmetric dimethylarginine (ADMA)–nitric oxide (NO) pathway. This review provides an overview of evidence from human studies supporting fetal programming of CVD, insight from animal models of cardiovascular programming and oxidative stress, impact of the l-arginine–ADMA–NO pathway in cardiovascular programming, the crosstalk between l-arginine metabolism and nutrient sensing signals, and application of reprogramming interventions to prevent the programming of CVD. A greater understanding of the mechanisms underlying cardiovascular programming is essential to developing early reprogramming interventions to combat the globally growing epidemic of CVD.
Collapse
|
30
|
The asymmetric dimethylarginine-mediated inhibition of nitric oxide in the rostral ventrolateral medulla contributes to regulation of blood pressure in hypertensive rats. Nitric Oxide 2017; 67:58-67. [PMID: 28392446 DOI: 10.1016/j.niox.2017.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 03/23/2017] [Accepted: 04/05/2017] [Indexed: 02/07/2023]
Abstract
Nitric oxide (NO) contributes to the central control of cardiovascular activity. The rostral ventrolateral medulla (RVLM) has been recognized as a pivotal region for maintaining basal blood pressure (BP) and sympathetic tone. It is reported that asymmetric dimethylarginine (ADMA), characterized as a cardiovascular risk marker, is an endogenous inhibitor of nitric oxide synthesis. The present was designed to determine the role of ADMA in the RVLM in the central control of BP in hypertensive rats. In Sprague Dawley (SD) rats, microinjection of ADMA into the RVLM dose-dependently increased BP, heart rate (HR), and renal sympathetic never activity (RSNA), but also reduced total NO production in the RVLM. In central angiotensin II (Ang II)-induced hypertensive rats and spontaneously hypertensive rat (SHR), the level of ADMA in the RVLM was increased and total NO production was decreased significantly, compared with SD rats treated vehicle infusion and WKY rats, respectively. These hypertensive rats also showed an increased protein level of protein arginine methyltransferases1 (PRMT1, which generates ADMA) and a decreased expression level of dimethylarginine dimethylaminohydrolases 1 (DDAH1, which degrades ADMA) in the RVLM. Furthermore, increased AMDA content and PRMT1 expression, and decreased levels of total NO production and DDAH1 expression in the RVLM in SHR were blunted by intracisternal infusion of the angiotensin II type 1 receptor (AT1R) blocker losartan. The current data indicate that the ADMA-mediated NO inhibition in the RVLM plays a critical role in involving in the central regulation of BP in hypertension, which may be associated with increased Ang II.
Collapse
|
31
|
Liu Z, Liu Y, Xu Q, Peng H, Tang Y, Yang T, Yu Z, Cheng G, Zhang G, Shi R. Critical role of vascular peroxidase 1 in regulating endothelial nitric oxide synthase. Redox Biol 2017; 12:226-232. [PMID: 28264790 PMCID: PMC5338721 DOI: 10.1016/j.redox.2017.02.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 02/26/2017] [Indexed: 12/19/2022] Open
Abstract
Vascular peroxidase 1 (VPO1) is a member of the peroxidase family which aggravates oxidative stress by producing hypochlorous acid (HOCl). Our previous study demonstrated that VPO1 plays a critical role in endothelial dysfunction through dimethylarginine dimethylaminohydrolase2 (DDAH2)/asymmetric Dimethylarginine (ADMA) pathway. Hereby we describe the regulatory role of VPO1 on endothelial nitric oxide synthase (eNOS) expression and activity in human umbilical vein endothelial cells (HUVECs). In HUVECs AngiotensinII (100 nM) treatment reduced Nitric Oxide (NO) production, decreased eNOS expression and activity, which were reversed by VPO1 siRNA. Knockdown of VPO1 also attenuated ADMA production and eNOS uncoupling while enhancing phosphorylated ser1177 eNOS expression level. Furthermore, HOCl stimulation was shown to directly induce ADMA production and eNOS uncoupling, decrease phosphorylated ser1177 eNOS expression. It also significantly suppressed eNOS expression and activity together with NO production. Therefore, VPO1 plays a vital role in regulating eNOS expression and activity via hydrogen peroxide (H2O2)-VPO1-HOCl pathway. Angiotensin II decreased eNOS expression and activity in HUVECs. VPO1 plays an important role in regulating eNOS expression and activity in HUVECs. VPO1 regulates eNOS expression and activity through VPO1/H2O2/HOCl pathway.
Collapse
Affiliation(s)
- Zhaoya Liu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yanbo Liu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qian Xu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Haiyang Peng
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yixin Tang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Tianlun Yang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zaixin Yu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Guangjie Cheng
- Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Guogang Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Ruizheng Shi
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
32
|
Vanhoutte PM, Shimokawa H, Feletou M, Tang EHC. Endothelial dysfunction and vascular disease - a 30th anniversary update. Acta Physiol (Oxf) 2017; 219:22-96. [PMID: 26706498 DOI: 10.1111/apha.12646] [Citation(s) in RCA: 620] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/27/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023]
Abstract
The endothelium can evoke relaxations of the underlying vascular smooth muscle, by releasing vasodilator substances. The best-characterized endothelium-derived relaxing factor (EDRF) is nitric oxide (NO) which activates soluble guanylyl cyclase in the vascular smooth muscle cells, with the production of cyclic guanosine monophosphate (cGMP) initiating relaxation. The endothelial cells also evoke hyperpolarization of the cell membrane of vascular smooth muscle (endothelium-dependent hyperpolarizations, EDH-mediated responses). As regards the latter, hydrogen peroxide (H2 O2 ) now appears to play a dominant role. Endothelium-dependent relaxations involve both pertussis toxin-sensitive Gi (e.g. responses to α2 -adrenergic agonists, serotonin, and thrombin) and pertussis toxin-insensitive Gq (e.g. adenosine diphosphate and bradykinin) coupling proteins. New stimulators (e.g. insulin, adiponectin) of the release of EDRFs have emerged. In recent years, evidence has also accumulated, confirming that the release of NO by the endothelial cell can chronically be upregulated (e.g. by oestrogens, exercise and dietary factors) and downregulated (e.g. oxidative stress, smoking, pollution and oxidized low-density lipoproteins) and that it is reduced with ageing and in the course of vascular disease (e.g. diabetes and hypertension). Arteries covered with regenerated endothelium (e.g. following angioplasty) selectively lose the pertussis toxin-sensitive pathway for NO release which favours vasospasm, thrombosis, penetration of macrophages, cellular growth and the inflammatory reaction leading to atherosclerosis. In addition to the release of NO (and EDH, in particular those due to H2 O2 ), endothelial cells also can evoke contraction of the underlying vascular smooth muscle cells by releasing endothelium-derived contracting factors. Recent evidence confirms that most endothelium-dependent acute increases in contractile force are due to the formation of vasoconstrictor prostanoids (endoperoxides and prostacyclin) which activate TP receptors of the vascular smooth muscle cells and that prostacyclin plays a key role in such responses. Endothelium-dependent contractions are exacerbated when the production of nitric oxide is impaired (e.g. by oxidative stress, ageing, spontaneous hypertension and diabetes). They contribute to the blunting of endothelium-dependent vasodilatations in aged subjects and essential hypertensive and diabetic patients. In addition, recent data confirm that the release of endothelin-1 can contribute to endothelial dysfunction and that the peptide appears to be an important contributor to vascular dysfunction. Finally, it has become clear that nitric oxide itself, under certain conditions (e.g. hypoxia), can cause biased activation of soluble guanylyl cyclase leading to the production of cyclic inosine monophosphate (cIMP) rather than cGMP and hence causes contraction rather than relaxation of the underlying vascular smooth muscle.
Collapse
Affiliation(s)
- P. M. Vanhoutte
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| | - H. Shimokawa
- Department of Cardiovascular Medicine; Tohoku University; Sendai Japan
| | - M. Feletou
- Department of Cardiovascular Research; Institut de Recherches Servier; Suresnes France
| | - E. H. C. Tang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
- School of Biomedical Sciences; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| |
Collapse
|
33
|
Tain YL, Hsu CN. Targeting on Asymmetric Dimethylarginine-Related Nitric Oxide-Reactive Oxygen Species Imbalance to Reprogram the Development of Hypertension. Int J Mol Sci 2016; 17:ijms17122020. [PMID: 27918455 PMCID: PMC5187820 DOI: 10.3390/ijms17122020] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/22/2016] [Accepted: 11/29/2016] [Indexed: 02/07/2023] Open
Abstract
Adult-onset diseases, including hypertension, can originate from early life, known as the developmental origins of health and disease (DOHaD). Because the developing kidney is vulnerable to early-life insults, renal programming is considered key in the developmental programming of hypertension. Asymmetric dimethylarginine (ADMA), an endogenous nitric oxide (NO) synthase inhibitor, can regulate the NO–reactive oxygen species (ROS) balance, and is involved in the development of hypertension. Reprogramming interventions aimed at NO-ROS balance can be protective in both genetic and developmentally programmed hypertension. Here we review several emergent themes of the DOHaD approach regarding the impact of ADMA-related NO-ROS imbalance on programmed hypertension. We focus on the kidney in the following areas: mechanistic insights to interpret programmed hypertension; the impact of ADMA-related NO-ROS imbalance in both genetic and acquired animal models of hypertension; alterations of the renal transcriptome in response to ADMA in the developing kidney; and reprogramming strategies targeting ADMA-related NO-ROS balance to prevent programmed hypertension.
Collapse
Affiliation(s)
- You-Lin Tain
- Departments of Pediatrics, College of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung 833, Taiwan.
- Institute for Translational Research in Biomedicine, College of Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, Kaohsiung 833, Taiwan.
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan.
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
34
|
Heimlich JB, Speed JS, O'Connor PM, Pollock JS, Townes TM, Meiler SE, Kutlar A, Pollock DM. Endothelin-1 contributes to the progression of renal injury in sickle cell disease via reactive oxygen species. Br J Pharmacol 2016; 173:386-95. [PMID: 26561980 DOI: 10.1111/bph.13380] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 10/16/2015] [Accepted: 10/25/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Endothelin-1 (ET-1) is increased in patients with sickle cell disease and may contribute to the development of sickle cell nephropathy. The current study was designed to determine whether ET-1 acting via the ETA receptor contributes to renal injury in a mouse model of sickle cell disease. EXPERIMENTAL APPROACH Adult, humanized HbSS (homozygous for sickle Hb) mice had increased ET-1 mRNA expression in both the cortex and the glomeruli compared with mice heterozygous for sickle and Hb A (HbAS controls). In the renal cortex, ETA receptor mRNA expression was also elevated in HbSS (sickle) mice although ETB receptor mRNA expression was unchanged. Ligand binding assays confirmed that sickle mice had increased ETA receptors in the renal vascular tissue when compared with control mice. KEY RESULTS In response to PKC stimulation, reactive oxygen species production by isolated glomeruli from HbSS sickle mice was increased compared with that from HbSA controls, an effect that was prevented by 1 week in vivo treatment with the selective ETA antagonist, ABT-627. Protein and nephrin excretion were both elevated in sickle mice, effects that were also significantly attenuated by ABT-627. Finally, ETA receptor antagonism caused a significant reduction in mRNA expression of NADPH oxidase subunits, which may contribute to nephropathy in sickle cell disease. CONCLUSIONS AND IMPLICATIONS These data support a novel role for ET-1 in the progression of sickle nephropathy, specifically via the ETA receptor, and suggest a potential role for ETA receptor antagonism in a treatment strategy.
Collapse
Affiliation(s)
- J Brett Heimlich
- 1epartment of Physiology, Augusta University, Augusta, GA 30912, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Morris RC, Schmidlin O, Sebastian A, Tanaka M, Kurtz TW. Vasodysfunction That Involves Renal Vasodysfunction, Not Abnormally Increased Renal Retention of Sodium, Accounts for the Initiation of Salt-Induced Hypertension. Circulation 2016; 133:881-93. [PMID: 26927006 DOI: 10.1161/circulationaha.115.017923] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- R Curtis Morris
- From the Departments of Medicine (R.C.M., O.S., A.S., M.T.) and Laboratory Medicine (T.W.K.), University of California, San Francisco.
| | - Olga Schmidlin
- From the Departments of Medicine (R.C.M., O.S., A.S., M.T.) and Laboratory Medicine (T.W.K.), University of California, San Francisco
| | - Anthony Sebastian
- From the Departments of Medicine (R.C.M., O.S., A.S., M.T.) and Laboratory Medicine (T.W.K.), University of California, San Francisco
| | - Masae Tanaka
- From the Departments of Medicine (R.C.M., O.S., A.S., M.T.) and Laboratory Medicine (T.W.K.), University of California, San Francisco
| | - Theodore W Kurtz
- From the Departments of Medicine (R.C.M., O.S., A.S., M.T.) and Laboratory Medicine (T.W.K.), University of California, San Francisco.
| |
Collapse
|
36
|
Hsu CP, Zhao JF, Lin SJ, Shyue SK, Guo BC, Lu TM, Lee TS. Asymmetric Dimethylarginine Limits the Efficacy of Simvastatin Activating Endothelial Nitric Oxide Synthase. J Am Heart Assoc 2016; 5:e003327. [PMID: 27091343 PMCID: PMC4843600 DOI: 10.1161/jaha.116.003327] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Asymmetric dimethylarginine (ADMA), an endogenous inhibitor of endothelial nitric oxide synthase (eNOS), is considered a risk factor for the pathogenesis of cardiovascular diseases. Simvastatin, a lipid‐lowering drug with other pleiotropic effects, has been widely used for treatment of cardiovascular diseases. However, little is known about the effect and underlying molecular mechanisms of ADMA on the effectiveness of simvastatin in the vascular system. Methods and Results We conducted a prospective cohort study to enroll 648 consecutive patients with coronary artery disease for a follow‐up period of 8 years. In patients with plasma ADMA level ≥0.49 μmol/L (a cut‐off value from receiver operating characteristic curve), statin treatment had no significant effect on cardiovascular events. We also conducted randomized, controlled studies using in vitro and in vivo models. In endothelial cells, treatment with ADMA (≥0.5 μmol/L) impaired simvastatin‐induced nitric oxide (NO) production, endothelial NO synthase (eNOS) phosphorylation, and angiogenesis. In parallel, ADMA markedly increased the activity of NADPH oxidase (NOX) and production of reactive oxygen species (ROS). The detrimental effects of ADMA on simvastatin‐induced NO production and angiogenesis were abolished by the antioxidant, N‐acetylcysteine, NOX inhibitor, or apocynin or overexpression of dimethylarginine dimethylaminohydrolase 2 (DDAH‐2). Moreover, in vivo, ADMA administration reduced Matrigel plug angiogenesis in wild‐type mice and decreased simvastatin‐induced eNOS phosphorylation in aortas of apolipoprotein E–deficient mice, but not endothelial DDAH‐2‐overexpressed aortas. Conclusions We conclude that ADMA may trigger NOX‐ROS signaling, which leads to restricting the simvastatin‐conferred protection of eNOS activation, NO production, and angiogenesis as well as the clinical outcome of cardiovascular events.
Collapse
Affiliation(s)
- Chiao-Po Hsu
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jin-Feng Zhao
- Department of Physiology, Genome Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Shing-Jong Lin
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Song-Kun Shyue
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Bei-Chia Guo
- Department of Physiology, Genome Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Tse-Min Lu
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzong-Shyuan Lee
- Department of Physiology, Genome Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
37
|
Javadzadeh A, Ghorbanihaghjo A, Mansouri S, Rashtchizadeh N. Asymmetric dimethylarginine and homocysteine in exudative age-related macular degeneration. JOURNAL OF ANALYTICAL RESEARCH IN CLINICAL MEDICINE 2015. [DOI: 10.15171/jarcm.2015.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
38
|
Spence JD, Urquhart BL, Bang H. Effect of renal impairment on atherosclerosis: only partially mediated by homocysteine. Nephrol Dial Transplant 2015; 31:937-44. [PMID: 26567910 DOI: 10.1093/ndt/gfv380] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 10/08/2015] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Cardiovascular risk and plasma total homocysteine (tHcy) are high in patients with renal failure. High tHcy may account for a substantial part of the increased risk. We assessed mediation by tHcy of the association of estimated glomerular filtration rate (eGFR CKD/EPI) with carotid total plaque area (TPA) and carotid stenosis. METHODS TPA and carotid stenosis were measured by ultrasound. Multiple linear regression was used to assess the effects of eGFR and/or tHcy after adjustment for age, sex, systolic blood pressure (SBP), smoking, LDL, HDL and weight. RESULTS Complete data were available for 1967 patients. eGFR decreased, and TPA and total stenosis increased linearly with age. After adjustment [age, sex, SBP, smoking (in pack years), low-density lipoprotein (LDL), high-density lipoprotein (HDL) and weight], eGFR and tHcy were independently associated with TPA (P < 0.01), but when both were added to the model, their significance was attenuated (P = 0.06 for eGFR, 0.03 for tHcy). Mediation analysis showed that tHcy seems to contribute to a significant mediation of the association of eGFR with TPA but not stenosis; after adjustment for the set of risk factors listed above, tHcy still demonstrated significant mediation on TPA (P = 0.03), but not on stenosis (P = 0.16). CONCLUSIONS tHcy accounts for a significant part, but not all of the effect of renal impairment on atherosclerosis. Other uremic toxins including metabolic products of the intestinal microbiome may explain residual effects of renal failure on atherosclerosis. Therapeutic approaches arising from that hypothesis are discussed.
Collapse
Affiliation(s)
- J David Spence
- Stroke Prevention and Atherosclerosis Research Centre, Robarts Research Institute, Western University, London, ON, Canada
| | - Bradley L Urquhart
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Heejung Bang
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA, USA
| |
Collapse
|
39
|
Gamboa JL, Pretorius M, Sprinkel KC, Brown NJ, Ikizler TA. Angiotensin converting enzyme inhibition increases ADMA concentration in patients on maintenance hemodialysis--a randomized cross-over study. BMC Nephrol 2015; 16:167. [PMID: 26494370 PMCID: PMC4618919 DOI: 10.1186/s12882-015-0162-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 10/12/2015] [Indexed: 02/07/2023] Open
Abstract
Background Endothelial dysfunction occurs in patients with end-stage renal disease (ESRD) and is associated with increased cardiovascular morbidity and mortality. Asymmetric dimethylarginine (ADMA) contributes to endothelial dysfunction in ESRD. In the general population, angiotensin-converting enzyme (ACE) inhibitors and angiotensin receptor blockers (ARBs) decrease ADMA levels, but no study has compared the effect of these drugs in patients with ESRD on maintenance hemodialysis (MHD). Methods We evaluated the effect of 1-week treatment with ramipril (5 mg/d), valsartan (160 mg/d), and placebo on ADMA levels in 15 patients on MHD in a double-blind, placebo-controlled, three x three cross-over study. Results We found that ADMA levels were increased at baseline and throughout the dialysis session during ramipril treatment (p < 0.001 compared to both, placebo and valsartan). Ramipril did not increase ADMA levels in a study of patients without ESRD, suggesting that factors related to ESRD or hemodialysis contribute to the ACE inhibitor-induced increase in ADMA. We have previously shown that ACE inhibition increases bradykinin (BK) levels during hemodialysis. We therefore evaluated the effect of bradykinin on ADMA production in A549 cells; a cell line that expresses BK receptors. Incubation with BK increased intracellular ADMA concentration through BK B2-receptor stimulation. Conclusion These data indicate that short-term ACE inhibition increases ADMA in patients on MHD whereas ARBs do not. In vitro studies further suggest that this may occur through BK-mediated increase in ADMA production during ACE inhibition. Trial registration Clinicaltrials.gov NCT00732069 August 6 2008 and NCT00607672 February 4 2008 Electronic supplementary material The online version of this article (doi:10.1186/s12882-015-0162-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jorge L Gamboa
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 2222 Pierce Avenue, 561-B PRB, Nashville, TN, 37232, USA.
| | - Mias Pretorius
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Katie C Sprinkel
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 2222 Pierce Avenue, 561-B PRB, Nashville, TN, 37232, USA.
| | - Nancy J Brown
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 2222 Pierce Avenue, 561-B PRB, Nashville, TN, 37232, USA.
| | - T Alp Ikizler
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
40
|
Porcelli V, Longo A, Palmieri L, Closs EI, Palmieri F. Asymmetric dimethylarginine is transported by the mitochondrial carrier SLC25A2. Amino Acids 2015; 48:427-36. [PMID: 26403849 DOI: 10.1007/s00726-015-2096-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/08/2015] [Indexed: 11/29/2022]
Abstract
Asymmetric dimethyl L-arginine (ADMA) is generated within cells and in mitochondria when proteins with dimethylated arginine residues are degraded. The aim of this study was to identify the carrier protein(s) that transport ADMA across the inner mitochondrial membrane. It was found that the recombinant, purified mitochondrial solute carrier SLC25A2 when reconstituted into liposomes efficiently transports ADMA in addition to its known substrates arginine, lysine, and ornithine and in contrast to the other known mitochondrial amino acid transporters SLC25A12, SLC25A13, SLC25A15, SLC25A18, SLC25A22, and SLC25A29. The widely expressed SLC25A2 transported ADMA across the liposomal membrane in both directions by both unidirectional transport and exchange against arginine or lysine. The SLC25A2-mediated ADMA transport followed first-order kinetics, was nearly as fast as the transport of the best SLC25A2 substrates known so far, and was highly specific as symmetric dimethylarginine (SDMA) was not transported at all. Furthermore, ADMA inhibited SLC25A2 activity with an inhibition constant of 0.38 ± 0.04 mM, whereas SDMA inhibited it poorly. We propose that a major function of SLC25A2 is to export ADMA from mitochondria missing the mitochondrial ADMA-metabolizing enzyme AGXT2. There is evidence that ADMA can also be imported into mitochondria, e.g., in kidney proximal tubulus cells, to be metabolized by AGXT2. SLC25A2 may also mediate this transport function.
Collapse
Affiliation(s)
- Vito Porcelli
- Laboratory of Biochemistry and Molecular Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona 4, 70125, Bari, Italy
| | - Antonella Longo
- Laboratory of Biochemistry and Molecular Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona 4, 70125, Bari, Italy
| | - Luigi Palmieri
- Laboratory of Biochemistry and Molecular Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona 4, 70125, Bari, Italy
| | - Ellen I Closs
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University, Obere Zahlbacher Strasse 67, 55101, Mainz, Germany
| | - Ferdinando Palmieri
- Laboratory of Biochemistry and Molecular Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona 4, 70125, Bari, Italy.
| |
Collapse
|
41
|
Ishima Y, Narisoko T, Kragh-Hansen U, Kotani S, Nakajima M, Otagiri M, Maruyama T. Nitration of indoxyl sulfate facilitates its cytotoxicity in human renal proximal tubular cells via expression of heme oxygenase-1. Biochem Biophys Res Commun 2015; 465:481-7. [PMID: 26277392 DOI: 10.1016/j.bbrc.2015.08.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 08/10/2015] [Indexed: 02/06/2023]
Abstract
Peroxynitrite, the reaction product of superoxide [Formula: see text] and nitric oxide (NO), nitrates tyrosine residues, unsaturated fatty acids, cyclic guanosine monophosphate and other phenolics. We report herein that indoxyl sulfate (IS) is also nitrated by peroxynitrite in vitro and forms 2-nitro-IS, as determined from spectral characteristics and (1)H-NMR. IS is one of the very important uremic toxins that accelerate the progression of chronic kidney disease via various mechanisms. However, cell viability experiments with human proximal tubular cells show that the cytotoxicity of 2-nitro-IS is several-fold higher than that of IS. The explanation for this finding seems to be that 2-nitro-IS induces a much more pronounced generation of intracellular reactive oxygen species (ROS) than IS. Results with inhibitors revealed that an organic anion transporter, several intracellular enzymes and nonprotein-bound iron ions are reasons for this finding. Most importantly, however, as detected by immunofluorescence and Western blotting, 2-nitro-IS induces the expression of heme oxygenase-1 and thereby the formation of ROS; most probably through the Fenton reaction. The final result of the increased amounts of ROS is death of the kidney cells. Thus, nitration of uremic toxins by peroxynitrite may help us to understand the initiation and progress of chronic kidney diseases.
Collapse
Affiliation(s)
- Yu Ishima
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto 862-0973, Japan; Center for Clinical Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto 862-0973, Japan
| | - Toru Narisoko
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto 862-0973, Japan
| | | | - Shunsuke Kotani
- Department of Organic Chemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto 862-0973, Japan
| | - Makoto Nakajima
- Department of Organic Chemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto 862-0973, Japan
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; Drug Delivery System Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto 862-0973, Japan; Center for Clinical Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto 862-0973, Japan.
| |
Collapse
|
42
|
Abstract
More is known about the epidemiology of drug-resistant hypertension than particular pathogenic factors and pathways. Several recurring themes, however, seem evident on using insight from epidemiology and general knowledge of the pathophysiology of hypertension. Specifically, 4 main pathways converge on drug resistance including sodium handling, sympathetic nervous system activation, endothelial dysfunction, and arterial stiffness. These factors, and the various pathways and elements contributing to them, are reviewed. In addition to describing how these factors exert their individual influences on resistant hypertension, several examples of how interactions between these factors, particularly in the case of chronic kidney disease, are included. At the conclusion of this review some thoughts are offered on additional mechanisms and areas for potential research.
Collapse
|
43
|
Morales Y, Nitzel DV, Price OM, Gui S, Li J, Qu J, Hevel JM. Redox Control of Protein Arginine Methyltransferase 1 (PRMT1) Activity. J Biol Chem 2015; 290:14915-26. [PMID: 25911106 DOI: 10.1074/jbc.m115.651380] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Indexed: 12/16/2022] Open
Abstract
Elevated levels of asymmetric dimethylarginine (ADMA) correlate with risk factors for cardiovascular disease. ADMA is generated by the catabolism of proteins methylated on arginine residues by protein arginine methyltransferases (PRMTs) and is degraded by dimethylarginine dimethylaminohydrolase. Reports have shown that dimethylarginine dimethylaminohydrolase activity is down-regulated and PRMT1 protein expression is up-regulated under oxidative stress conditions, leading many to conclude that ADMA accumulation occurs via increased synthesis by PRMTs and decreased degradation. However, we now report that the methyltransferase activity of PRMT1, the major PRMT isoform in humans, is impaired under oxidative conditions. Oxidized PRMT1 displays decreased activity, which can be rescued by reduction. This oxidation event involves one or more cysteine residues that become oxidized to sulfenic acid (-SOH). We demonstrate a hydrogen peroxide concentration-dependent inhibition of PRMT1 activity that is readily reversed under physiological H2O2 concentrations. Our results challenge the unilateral view that increased PRMT1 expression necessarily results in increased ADMA synthesis and demonstrate that enzymatic activity can be regulated in a redox-sensitive manner.
Collapse
Affiliation(s)
- Yalemi Morales
- From the Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322
| | - Damon V Nitzel
- From the Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322
| | - Owen M Price
- From the Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322
| | - Shanying Gui
- From the Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322
| | - Jun Li
- the Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York 14260, and the New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York 14203
| | - Jun Qu
- the Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York 14260, and the New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York 14203
| | - Joan M Hevel
- From the Department of Chemistry and Biochemistry, Utah State University, Logan, Utah 84322,
| |
Collapse
|
44
|
Armando I, Konkalmatt P, Felder RA, Jose PA. The renal dopaminergic system: novel diagnostic and therapeutic approaches in hypertension and kidney disease. Transl Res 2015; 165:505-11. [PMID: 25134060 PMCID: PMC4305499 DOI: 10.1016/j.trsl.2014.07.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 07/17/2014] [Accepted: 07/19/2014] [Indexed: 12/15/2022]
Abstract
Salt sensitivity of blood pressure, whether in hypertensive or normotensive subjects, is associated with increased cardiovascular risk and overall mortality. Salt sensitivity can be treated by reducing NaCl consumption. However, decreasing salt intake in some may actually increase cardiovascular risk, including an increase in blood pressure, that is, inverse salt sensitivity. Several genes have been associated with salt sensitivity and inverse salt sensitivity. Some of these genes encode proteins expressed in the kidney that are needed to excrete a sodium load, for example, dopamine receptors and their regulators, G protein-coupled receptor kinase 4 (GRK4). We review here research in this field that has provided several translational opportunities, ranging from diagnostic tests to gene therapy, such as (1) a test in renal proximal tubule cells isolated from the urine of humans that may determine the salt-sensitive phenotype by analyzing the recruitment of dopamine D1 receptors to the plasma membrane; (2) the presence of common GRK4 gene variants that are not only associated with hypertension but may also be predictive of the response to antihypertensive therapy; (3) genetic testing for polymorphisms of the dopamine D2 receptor that may be associated with hypertension and inverse salt sensitivity and may increase the susceptibility to chronic kidney disease because of loss of the antioxidant and anti-inflammatory effects of the renal dopamine D2 receptor, and (4) in vivo renal selective amelioration of renal tubular genetic defects by a gene transfer approach, using adeno-associated viral vectors introduced to the kidney by retrograde ureteral infusion.
Collapse
Affiliation(s)
- Ines Armando
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Prasad Konkalmatt
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Robin A Felder
- Department of Pathology, The University of Virginia School of Medicine, Charlottesville, VA
| | - Pedro A Jose
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD; Department of Physiology, University of Maryland School of Medicine, Baltimore, MD.
| |
Collapse
|
45
|
Eelen G, de Zeeuw P, Simons M, Carmeliet P. Endothelial cell metabolism in normal and diseased vasculature. Circ Res 2015; 116:1231-44. [PMID: 25814684 PMCID: PMC4380230 DOI: 10.1161/circresaha.116.302855] [Citation(s) in RCA: 446] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/25/2015] [Indexed: 12/25/2022]
Abstract
Higher organisms rely on a closed cardiovascular circulatory system with blood vessels supplying vital nutrients and oxygen to distant tissues. Not surprisingly, vascular pathologies rank among the most life-threatening diseases. At the crux of most of these vascular pathologies are (dysfunctional) endothelial cells (ECs), the cells lining the blood vessel lumen. ECs display the remarkable capability to switch rapidly from a quiescent state to a highly migratory and proliferative state during vessel sprouting. This angiogenic switch has long been considered to be dictated by angiogenic growth factors (eg, vascular endothelial growth factor) and other signals (eg, Notch) alone, but recent findings show that it is also driven by a metabolic switch in ECs. Furthermore, these changes in metabolism may even override signals inducing vessel sprouting. Here, we review how EC metabolism differs between the normal and dysfunctional/diseased vasculature and how it relates to or affects the metabolism of other cell types contributing to the pathology. We focus on the biology of ECs in tumor blood vessel and diabetic ECs in atherosclerosis as examples of the role of endothelial metabolism in key pathological processes. Finally, current as well as unexplored EC metabolism-centric therapeutic avenues are discussed.
Collapse
Affiliation(s)
- Guy Eelen
- From the Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium (G.E., P.d.Z., P.C.); Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium (G.E., P.d.Z., P.C.); Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, New Haven, CT (M.S.); and Department of Cell Biology, Yale University School of Medicine, New Haven, CT (M.S.)
| | - Pauline de Zeeuw
- From the Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium (G.E., P.d.Z., P.C.); Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium (G.E., P.d.Z., P.C.); Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, New Haven, CT (M.S.); and Department of Cell Biology, Yale University School of Medicine, New Haven, CT (M.S.)
| | - Michael Simons
- From the Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium (G.E., P.d.Z., P.C.); Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium (G.E., P.d.Z., P.C.); Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, New Haven, CT (M.S.); and Department of Cell Biology, Yale University School of Medicine, New Haven, CT (M.S.)
| | - Peter Carmeliet
- From the Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium (G.E., P.d.Z., P.C.); Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium (G.E., P.d.Z., P.C.); Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, New Haven, CT (M.S.); and Department of Cell Biology, Yale University School of Medicine, New Haven, CT (M.S.).
| |
Collapse
|
46
|
Shing CM, Fassett RG, Peake JM, Coombes JS. Voluntary exercise decreases atherosclerosis in nephrectomised ApoE knockout mice. PLoS One 2015; 10:e0120287. [PMID: 25799529 PMCID: PMC4370520 DOI: 10.1371/journal.pone.0120287] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 02/02/2015] [Indexed: 11/21/2022] Open
Abstract
Cardiovascular disease is the main cause of morbidity and mortality in patients with kidney disease. The effectiveness of exercise for cardiovascular disease that is accelerated by the presence of chronic kidney disease remains unknown. The present study utilized apolipoprotein E knockout mice with 5/6 nephrectomy as a model of combined kidney disease and cardiovascular disease to investigate the effect of exercise on aortic plaque formation, vascular function and systemic inflammation. Animals were randomly assigned to nephrectomy or control and then to either voluntary wheel running exercise or sedentary. Following 12-weeks, aortic plaque area was significantly (p<0.05, d=1.2) lower in exercising nephrectomised mice compared to sedentary nephrectomised mice. There was a strong, negative correlation between average distance run each week and plaque area in nephrectomised and control mice (r=–0.76, p=0.048 and r=–0.73, p=0.062; respectively). In vitro aortic contraction and endothelial-independent and endothelial-dependent relaxation were not influenced by exercise (p>0.05). Nephrectomy increased IL-6 and TNF-α concentrations compared with control mice (p<0.001 and p<0.05, respectively), while levels of IL-10, MCP-1 and MIP-1α were not significantly influenced by nephrectomy or voluntary exercise (p>0.05). Exercise was an effective non-pharmacologic approach to slow cardiovascular disease in the presence of kidney disease in the apolipoprotein E knockout mouse.
Collapse
Affiliation(s)
- Cecilia M. Shing
- School of Health Sciences, University of Tasmania, Launceston, Tasmania, Australia
- * E-mail:
| | - Robert G. Fassett
- School of Human Movement Studies, University of Queensland, Brisbane, Queensland, Australia
| | - Jonathan M. Peake
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Jeff S. Coombes
- School of Human Movement Studies, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
47
|
Yang S, Yang Y, Yu P, Yang J, Jiang X, Villar VAM, Sibley DR, Jose PA, Zeng C. Dopamine D1 and D5 receptors differentially regulate oxidative stress through paraoxonase 2 in kidney cells. Free Radic Res 2015; 49:397-410. [PMID: 25740199 DOI: 10.3109/10715762.2015.1006215] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The renal dopaminergic system plays an important role in the pathogenesis of hypertension. Dopamine D1-like receptors (D1R and D5R) decrease reactive oxygen species (ROS) production via inhibition of pro-oxidant enzymes such as NADPH oxidase. Paraoxonase 2 (PON2) is also involved in the inhibition of NADPH oxidase activity. Therefore, we tested the hypothesis that D1R and D5R inhibit ROS production by increasing the expression of PON2, including those in membrane microdomains. METHODS AND RESULTS PON2 colocalized with D1R and D5R in mouse renal proximal tubules (RPTs), human RPT (hRPT) cells, and HEK293 cells heterologously expressing human D1R (HEK-hD1R) or D5R (HEK-hD5R). Fenoldopam, an agonist for both D1R and D5R, increased PON2 co-immunoprecipitation with D1R and D5R in HEK-hD1R and HEK-hD5R cells, respectively. Silencing PON2 increased ROS production and NADPH oxidase activity, and impaired the inhibitory effect of fenoldopam. Fenoldopam increased PON2 protein in both lipid rafts (LRs) and non-LRs in HEK-hD1R cells, but only in non-LRs in HEK-hD5R and hRPT cells. Long-term (hrs) fenoldopam stimulation increased PON2 protein in a time-dependent manner in HEK-hD5R, but not in HEK-hD1R cells. Because the effects of fenoldopam on non-LR and total PON2 expressions were similar in HEK-hD5R and hRPT cells, additional studies were performed to determine the relationship between D5R and PON2. Renal PON2 protein was decreased in D5(-/-) mice. In hRPT cells, silencing D5R decreased PON2 expression and increased ROS production. CONCLUSIONS We conclude that D1-like receptors inhibit ROS production by altering PON2 distribution in membrane microdomains in the short-term, and by increasing PON2 expression in the long-term.
Collapse
Affiliation(s)
- S Yang
- Department of Cardiology, Daping Hospital, The Third Military Medical University , Chongqing , P. R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Seibert F, Behrendt C, Pagonas N, Bauer F, Kiziler F, Zidek W, Westhoff T. Prediction of Cardiovascular Events After Renal Transplantation. Transplant Proc 2015; 47:388-93. [DOI: 10.1016/j.transproceed.2014.12.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/25/2014] [Accepted: 12/31/2014] [Indexed: 01/08/2023]
|
49
|
Luo Z, Aslam S, Welch WJ, Wilcox CS. Activation of nuclear factor erythroid 2-related factor 2 coordinates dimethylarginine dimethylaminohydrolase/PPAR-γ/endothelial nitric oxide synthase pathways that enhance nitric oxide generation in human glomerular endothelial cells. Hypertension 2015; 65:896-902. [PMID: 25691623 DOI: 10.1161/hypertensionaha.114.04760] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Dimethylarginine dimethylaminohydrolase (DDAH) degrades asymmetric dimethylarginine, which inhibits nitric oxide (NO) synthase (NOS). Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcriptional factor that binds to antioxidant response elements and transcribes many antioxidant genes. Because the promoters of the human DDAH-1 and DDAH-2, endothelial NOS (eNOS) and PPAR-γ genes contain 2 to 3 putative antioxidant response elements, we hypothesized that they were regulated by Nrf2/antioxidant response element. Incubation of human renal glomerular endothelial cells with the Nrf2 activator tert-butylhydroquinone (20 μmol·L(-1)) significantly (P<0.05) increased NO and activities of NOS and DDAH and decreased asymmetric dimethylarginine. It upregulated genes for hemoxygenase-1, eNOS, DDAH-1, DDAH-2, and PPAR-γ and partitioned Nrf2 into the nucleus. Knockdown of Nrf2 abolished these effects. Nrf2 bound to one antioxidant response element on DDAH-1 and DDAH-2 and PPAR-γ promoters but not to the eNOS promoter. An increased eNOS and phosphorylated eNOS (P-eNOSser-1177) expression with tert-butylhydroquinone was prevented by knockdown of PPAR-γ. Expression of Nrf2 was reduced by knockdown of PPAR-γ, whereas PPAR-γ was reduced by knockdown of Nrf2, thereby demonstrating 2-way positive interactions. Thus, Nrf2 transcribes HO-1 and other genes to reduce reactive oxygen species, and DDAH-1 and DDAH-2 to reduce asymmetric dimethylarginine and PPAR-γ to increase eNOS and its phosphorylation and activity thereby coordinating 3 pathways that enhance endothelial NO generation.
Collapse
Affiliation(s)
- Zaiming Luo
- From the Division of Nephrology and Hypertension and Hypertension, Kidney and Vascular Research Center, Georgetown University, Washington, DC
| | - Shakil Aslam
- From the Division of Nephrology and Hypertension and Hypertension, Kidney and Vascular Research Center, Georgetown University, Washington, DC
| | - William J Welch
- From the Division of Nephrology and Hypertension and Hypertension, Kidney and Vascular Research Center, Georgetown University, Washington, DC
| | - Christopher S Wilcox
- From the Division of Nephrology and Hypertension and Hypertension, Kidney and Vascular Research Center, Georgetown University, Washington, DC.
| |
Collapse
|
50
|
Ware KM, Vance JC, Muni N, Hebert LA, Satoskar AA, Nadasdy G, Ivanov I, Nadasdy T, Rovin BH, Brodsky SV. Oral warfarin and the thrombin inhibitor dabigatran increase blood pressure in rats: hidden danger of anticoagulants? Am J Hypertens 2015; 28:182-9. [PMID: 25023204 DOI: 10.1093/ajh/hpu129] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Hypertension is a common comorbidity in patients with chronic kidney disease (CKD). We reported earlier that oral anticoagulants, including warfarin and dabigatran, may induce acute kidney injury. No effects of oral anticoagulants on blood pressure (BP) have been previously reported. The aim of this study was to examine in detail the relationship of anticoagulant therapy and BP in rats. METHODS Sham-operated and 5/6 nephrectomy rats were treated with different doses of oral anticoagulants (warfarin and dabigatran), superoxide scavenger N-acetylcysteine (NAC), vitamin K, and protease activated receptor 1 (PAR-1) inhibitor SCH79797. BP was measured by a tail cuff daily. RESULTS Warfarin and dabigatran both increased systolic BP in sham-operated and 5/6 nephrectomy rats in a dose-dependent manner. SCH79797 also increased systolic BP in a dose-dependent manner. Vitamin K prevented warfarin-induced increase in BP but did not affect BP when administered alone. NAC delayed the warfarin-associated increase in BP. Warfarin effects on BP were similar in 5/6 nephrectomy rats with different CKD stages. CONCLUSIONS Both warfarin and dabigatran increase systolic BP in rats. The mechanism of this effect is not clear, but our data suggest that it is related to decreased thrombin activity associated with anticoagulant treatment. The superoxide scavenger NAC delayed, but did not prevent, warfarin-induced hypertension.
Collapse
Affiliation(s)
- Kyle M Ware
- Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Jay C Vance
- Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Navin Muni
- Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Lee A Hebert
- Department of Medicine, The Ohio State University, Columbus, Ohio
| | | | - Gyongyi Nadasdy
- Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Iouri Ivanov
- Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Tibor Nadasdy
- Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Brad H Rovin
- Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Sergey V Brodsky
- Department of Pathology, The Ohio State University, Columbus, Ohio;
| |
Collapse
|