151
|
Balcilar C, Özakça I, Altan VM. Contribution of Rho-kinase and Adenosine Monophosphate-Activated Protein Kinase Signaling Pathways to Endothelium-Derived Contracting Factors Responses. Turk J Pharm Sci 2017; 14:207-212. [PMID: 32454615 DOI: 10.4274/tjps.26349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/10/2017] [Indexed: 12/01/2022]
Abstract
Vascular tonus is controlled by endothelium-derived relaxing factor (EDRF), endothelium-derived hyperpolarizing factor (EDHF) and endothelium-derived contracting factor (EDCF) under physiological circumstances. In pathological conditions, impairment of endothelium-derived relaxation can be caused by both decrease in EDRF release and increase in EDCF release. The increase in EDCF is observed with diseases such as hypertension and diabetes. The contribution of Rho-kinase and activated protein kinase (AMPK), which have opposite effects, to the increased EDCF responses was investigated. Rho-kinases are the effectors of Rho which is one of the small guanosine triphosphate-binding proteins. They increase cytosolic Ca+2 concentration and cause vascular smooth muscle to contract, keeping myosin light chain (MLC) in phosphorylated state by affecting myosin phosphatase target subunit which dephosphorylates the MLC. The activities of Rho-kinases increase with the increase of EDCF function. AMPK is the energy sensor of the cell. It provides a vasculoprotective effect by causing endothelium-dependent and endothelium-independent relaxation in smooth muscle. In contrast to Rho-kinase pathway activity, AMPK pathway activity decreases with diseases in which the EDCF function increases. In cases such as diabetes and hypertension that endothelial function impairs toward vasocontraction, it is considered that evaluating Rho-kinase and AMPK pathways which mediate contraction and relaxation in vascular smooth muscle respectively, would provide clues on choosing therapeutic target for pathologies in which endothelial dysfunction is observed.
Collapse
Affiliation(s)
- Cennet Balcilar
- Ankara University, Faculty Of Pharmacy, Department Of Pharmacology, Ankara, Turkey
| | - Işıl Özakça
- Ankara University, Faculty Of Pharmacy, Department Of Pharmacology, Ankara, Turkey
| | - Vecdi Melih Altan
- Ankara University, Faculty Of Pharmacy, Department Of Pharmacology, Ankara, Turkey
| |
Collapse
|
152
|
Mahmoud AM, Wilkinson FL, McCarthy EM, Moreno-Martinez D, Langford-Smith A, Romero M, Duarte J, Alexander MY. Endothelial microparticles prevent lipid-induced endothelial damage via Akt/eNOS signaling and reduced oxidative stress. FASEB J 2017; 31:4636-4648. [PMID: 28687612 PMCID: PMC5714503 DOI: 10.1096/fj.201601244rr] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 06/27/2017] [Indexed: 12/31/2022]
Abstract
Endothelial microparticles (EMPs) are endothelium-derived submicron vesicles that are released in response to diverse stimuli and are elevated in cardiovascular disease, which is correlated with risk factors. This study investigates the effect of EMPs on endothelial cell function and dysfunction in a model of free fatty acid (FFA) palmitate-induced oxidative stress. EMPs were generated from TNF-α-stimulated HUVECs and quantified by using flow cytometry. HUVECs were treated with and without palmitate in the presence or absence of EMPs. EMPs were found to carry functional eNOS and to protect against oxidative stress by positively regulating eNOS/Akt signaling, which restored NO production, increased superoxide dismutase and catalase, and suppressed NADPH oxidase and reactive oxygen species (ROS) production, with the involvement of NF-erythroid 2-related factor 2 and heme oxygenase-1. Conversely, under normal conditions, EMPs reduced NO release and increased ROS and redox-sensitive marker expression. In addition, functional assays using EMP-treated mouse aortic rings that were performed under homeostatic conditions demonstrated a decline in endothelium-dependent vasodilatation, but restored the functional response under lipid-induced oxidative stress. These data indicate that EMPs harbor functional eNOS and potentially play a role in the feedback loop of damage and repair during homeostasis, but are also effective in protecting against FFA-induced oxidative stress; thus, EMP function is reflected by the microenvironment.-Mahmoud, A. M., Wilkinson, F. L., McCarthy, E. M., Moreno-Martinez, D., Langford-Smith, A., Romero, M., Duarte, J., Alexander, M. Y. Endothelial microparticles prevent lipid-induced endothelial damage via Akt/eNOS signaling and reduced oxidative stress.
Collapse
Affiliation(s)
- Ayman M Mahmoud
- Healthcare Science Research Centre, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom.,Manchester Academic Health Science Centre, Manchester, United Kingdom.,Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, Beni Suef, Egypt
| | - Fiona L Wilkinson
- Healthcare Science Research Centre, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom.,Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Eoghan M McCarthy
- Healthcare Science Research Centre, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom.,Manchester Academic Health Science Centre, Manchester, United Kingdom.,Centre for Musculoskeletal Research, Institute of Inflammation and Repair, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom.,Musculoskeletal Biomedical Research Unit, National Institute for Health Research Manchester, Central Manchester University Hospital NHS Foundation Trust, Manchester, United Kingdom
| | - Daniel Moreno-Martinez
- Healthcare Science Research Centre, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom.,Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Alexander Langford-Smith
- Healthcare Science Research Centre, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom.,Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Miguel Romero
- Department of Pharmacology, School of Pharmacy, University of Granada, Granada, Spain
| | - Juan Duarte
- Instituto de Investigación Biosanitaria de Granada, Granada, Spain
| | - M Yvonne Alexander
- Healthcare Science Research Centre, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom; .,Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
153
|
Arnal JF, Lenfant F, Metivier R, Flouriot G, Henrion D, Adlanmerini M, Fontaine C, Gourdy P, Chambon P, Katzenellenbogen B, Katzenellenbogen J. Membrane and Nuclear Estrogen Receptor Alpha Actions: From Tissue Specificity to Medical Implications. Physiol Rev 2017; 97:1045-1087. [DOI: 10.1152/physrev.00024.2016] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/19/2016] [Accepted: 01/18/2017] [Indexed: 12/22/2022] Open
Abstract
Estrogen receptor alpha (ERα) has been recognized now for several decades as playing a key role in reproduction and exerting functions in numerous nonreproductive tissues. In this review, we attempt to summarize the in vitro studies that are the basis of our current understanding of the mechanisms of action of ERα as a nuclear receptor and the key roles played by its two activation functions (AFs) in its transcriptional activities. We then depict the consequences of the selective inactivation of these AFs in mouse models, focusing on the prominent roles played by ERα in the reproductive tract and in the vascular system. Evidence has accumulated over the two last decades that ERα is also associated with the plasma membrane and activates non-nuclear signaling from this site. These rapid/nongenomic/membrane-initiated steroid signals (MISS) have been characterized in a variety of cell lines, and in particular in endothelial cells. The development of selective pharmacological tools that specifically activate MISS and the generation of mice expressing an ERα protein impeded for membrane localization have begun to unravel the physiological role of MISS in vivo. Finally, we discuss novel perspectives for the design of tissue-selective ER modulators based on the integration of the physiological and pathophysiological roles of MISS actions of estrogens.
Collapse
Affiliation(s)
- Jean-Francois Arnal
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Françoise Lenfant
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Raphaël Metivier
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Gilles Flouriot
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Daniel Henrion
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Marine Adlanmerini
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Coralie Fontaine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Pierre Gourdy
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Pierre Chambon
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Benita Katzenellenbogen
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - John Katzenellenbogen
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| |
Collapse
|
154
|
Pierce RW, Giuliano JS, Pober JS. Endothelial Cell Function and Dysfunction in Critically Ill Children. Pediatrics 2017; 140:peds.2017-0355. [PMID: 28759412 PMCID: PMC9923607 DOI: 10.1542/peds.2017-0355] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/20/2017] [Indexed: 11/24/2022] Open
Abstract
Endothelial cells (ECs) line the lumen of the entire vascular system and actively regulate blood flow; maintain blood fluidity; control water, solute, and macromolecular transfer between blood and tissue; and modulate circulating immune cell recruitment and activation. These vital functions, combined with the broad anatomic distribution of ECs, implicate them in all forms of critical illness. The present article discusses how ECs adapt and break down during the course of critical illness. We first review the biology of ECs, highlighting the vascular segmental differences and their specific roles in the maintenance of homeostasis. We then discuss how ECs acquire new functions to restore local and systemic homeostasis (activation) as well as how breakdowns in EC functions (dysfunction) contribute to local and systemic pathologic responses, with clinical correlations. Lastly, how these processes have been studied in critically ill children is discussed.
Collapse
Affiliation(s)
- Richard W. Pierce
- Departments of Pediatrics and,Address correspondence to Richard W. Pierce, MD, MS, Department of Pediatrics, Section of Critical Care Medicine, Yale University, 333 Cedar St, PO Box 208064, New Haven, CT 06520. E-mail:
| | | | | |
Collapse
|
155
|
Peiró C, Lorenzo Ó, Carraro R, Sánchez-Ferrer CF. IL-1β Inhibition in Cardiovascular Complications Associated to Diabetes Mellitus. Front Pharmacol 2017; 8:363. [PMID: 28659798 PMCID: PMC5468794 DOI: 10.3389/fphar.2017.00363] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 05/26/2017] [Indexed: 01/15/2023] Open
Abstract
Diabetes mellitus (DM) is a chronic disease that affects nowadays millions of people worldwide. In adults, type 2 diabetes mellitus (T2DM) accounts for the majority of all diagnosed cases of diabetes. The course of the T2DM is characterized by insulin resistance and a progressive loss of β-cell mass. DM is associated with a number of related complications, among which cardiovascular complications and atherosclerosis are the main cause of morbidity and mortality in patients suffering from the disease. DM is acknowledged as a low-grade chronic inflammatory state characterized by the over-secretion of pro-inflammatory cytokines, including interleukin (IL)-1β, which reinforce inflammatory signals thus contributing to the development of complications. In this context, the pharmacological approaches to treat diabetes should not only correct hyperglycaemia, but also attenuate inflammation and prevent the development of metabolic and cardiovascular complications. Over the last years, novel biological drugs have been developed to antagonize the pathophysiological actions of IL-1β. The drugs currently used in clinical practice are anakinra, a recombinant form of the naturally occurring IL-1 receptor antagonist, the soluble decoy receptor rilonacept and the monoclonal antibodies canakinumab and gevokizumab. This review will summarize the main experimental and clinical findings obtained with pharmacological IL-1β inhibitors in the context of the cardiovascular complications of DM, and discuss the perspectives of IL-1β inhibitors as novel therapeutic tools for treating these patients.
Collapse
Affiliation(s)
- Concepción Peiró
- Department of Pharmacology, School of Medicine, Universidad Autónoma de MadridMadrid, Spain.,Instituto de Investigación Sanitaria Hospital Universitario de La Paz (IdiPAZ)Madrid, Spain
| | - Óscar Lorenzo
- Department of Medicine, School of Medicine, Universidad Autónoma de MadridMadrid, Spain.,Instituto de Investigación Sanitaria Fundación Jiménez DíazMadrid, Spain
| | - Raffaele Carraro
- Department of Medicine, School of Medicine, Universidad Autónoma de MadridMadrid, Spain.,Service of Endocrinology, Hospital de La PrincesaMadrid, Spain.,Instituto de Investigación Sanitaria Hospital de La PrincesaMadrid, Spain
| | - Carlos F Sánchez-Ferrer
- Department of Pharmacology, School of Medicine, Universidad Autónoma de MadridMadrid, Spain.,Instituto de Investigación Sanitaria Hospital Universitario de La Paz (IdiPAZ)Madrid, Spain
| |
Collapse
|
156
|
Sex differences in micro- and macro-vascular complications of diabetes mellitus. Clin Sci (Lond) 2017; 131:833-846. [PMID: 28424377 DOI: 10.1042/cs20160998] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 01/24/2017] [Accepted: 01/25/2017] [Indexed: 12/14/2022]
Abstract
Vascular complications are a leading cause of morbidity and mortality in both men and women with type 1 (T1DM) or type 2 (T2DM) diabetes mellitus, however the prevalence, progression and pathophysiology of both microvascular (nephropathy, neuropathy and retinopathy) and macrovascular [coronary heart disease (CHD), myocardial infarction, peripheral arterial disease (PAD) and stroke] disease are different in the two sexes. In general, men appear to be at a higher risk for diabetic microvascular complications, while the consequences of macrovascular complications may be greater in women. Interestingly, in the absence of diabetes, women have a far lower risk of either micro- or macro-vascular disease compared with men for much of their lifespan. Thus, the presence of diabetes confers greater risk for vascular complications in women compared with men and some of the potential reasons, including contribution of sex hormones and sex-specific risk factors are discussed in this review. There is a growing body of evidence that sex hormones play an important role in the regulation of cardiovascular function. While estrogens are generally considered to be cardioprotective and androgens detrimental to cardiovascular health, recent findings challenge these assumptions and demonstrate diversity and complexity of sex hormone action on target tissues, especially in the setting of diabetes. While some progress has been made toward understanding the underlying mechanisms of sex differences in the pathophysiology of diabetic vascular complications, many questions and controversies remain. Future research leading to understanding of these mechanisms may contribute to personalized- and sex-specific treatment for diabetic micro- and macro-vascular disease.
Collapse
|
157
|
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
|
158
|
Wang ZR, Liu HB, Sun YY, Hu QQ, Li YX, Zheng WW, Yu CJ, Li XY, Wu MM, Song BL, Mu JJ, Yuan ZY, Zhang ZR, Ma HP. Dietary salt blunts vasodilation by stimulating epithelial sodium channels in endothelial cells from salt-sensitive Dahl rats. Br J Pharmacol 2017; 175:1305-1317. [PMID: 28409833 DOI: 10.1111/bph.13817] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/26/2017] [Accepted: 04/04/2017] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Our recent studies show that the reduced activity of epithelial sodium channels (ENaC) in endothelial cells accounts for the adaptation of vasculature to salt in Sprague-Dawley rats. The present study examines a hypothesis that enhanced ENaC activity mediates the loss of vasorelaxation in Dahl salt-sensitive (SS) rats. EXPERIMENTAL APPROACH We used the cell-attached patch-clamp technique to record ENaC activity in split-open mesenteric arteries. Western blot and immunofluorescence staining were used to evaluate the levels of aldosterone, ENaC, eNOS and NO. Blood pressure was measured with the tail-cuff method and the artery relaxation was measured with the wire myograph assay. KEY RESULTS High-salt (HS) diet significantly increased plasma aldosterone and ENaC activity in the endothelial cells of Dahl SS rats. The endothelium-dependent artery relaxation was blunted by HS challenge in these rats. Amiloride, a potent blocker of ENaC, increased both phosphorylated eNOS and NO and therefore prevented the HS-induced loss of vasorelaxation. As, in SS rats, endogenous aldosterone was already elevated by HS challenge, exogenous aldosterone did not further elevate ENaC activity in the rats fed with HS. Eplerenone, a mineralocorticoid receptor antagonist, attenuated the effects of HS on both ENaC activity and artery relaxation. CONCLUSIONS AND IMPLICATIONS These data suggest that HS diet blunts artery relaxation and causes hypertension via a pathway associated with aldosterone-dependent activation of ENaC in endothelial cells. This pathway provides one of the mechanisms by which HS causes hypertension in Dahl SS rats. LINKED ARTICLES This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.
Collapse
Affiliation(s)
- Zi-Rui Wang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Hui-Bin Liu
- Department of Clinical Pharmacy, Institute of Clinical Pharmacy, the 2nd Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ying-Ying Sun
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing-Qing Hu
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Yu-Xia Li
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Wei-Wan Zheng
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Chang-Jiang Yu
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Xin-Yuan Li
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Ming-Ming Wu
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Bin-Lin Song
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| | - Jian-Jun Mu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Key Laboratory of Molecular Cardiology, Xi'an, China
| | - Zu-Yi Yuan
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Key Laboratory of Molecular Cardiology, Xi'an, China
| | - Zhi-Ren Zhang
- Departments of Cardiology and Clinical Pharmacy, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China.,Department of Clinical Pharmacy, Institute of Clinical Pharmacy, the 2nd Affiliated Hospital, Harbin Medical University, Harbin, China
| | - He-Ping Ma
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
159
|
Ahmad KA, Yuan Yuan D, Nawaz W, Ze H, Zhuo CX, Talal B, Taleb A, Mais E, Qilong D. Antioxidant therapy for management of oxidative stress induced hypertension. Free Radic Res 2017; 51:428-438. [PMID: 28427291 DOI: 10.1080/10715762.2017.1322205] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hypertension is considered as the most common risk factor for cardiovascular diseases, also is regarded as a leading cause of the mortality and morbidity worldwide. The mechanisms underlying the pathological process of hypertension are not completely explained. However, there is growing evidence that increased oxidative stress plays an important role in the pathophysiology of hypertension. Several preclinical studies and clinical trials have indicated that antioxidant therapy is important for management of hypertension, using antioxidants compounds such as alpha tocopherol (Vit E) and ascorbic acid (Vit C), polyphenols with others and some antihypertensive drugs that are now in clinical use (e.g. ACEIs, ARBs, novel B-blockers, dihydropyridine CCBs) which have antioxidative pleiotropic effects. The purpose of this review is to highlight the importance of antioxidant therapy for management of oxidative stress induced hypertension. Furthermore, we review the current knowledge in the oxidative stress and its significance in hypertension.
Collapse
Affiliation(s)
- Khalil Ali Ahmad
- a Department of Pharmacology, School of Pharmacy , China Pharmaceutical University , Nanjing , China
| | - Dai Yuan Yuan
- a Department of Pharmacology, School of Pharmacy , China Pharmaceutical University , Nanjing , China
| | - Waqas Nawaz
- b School of Basic Medicine and Clinical Pharmacy , China Pharmaceutical University , Nanjing , China
| | - Hong Ze
- a Department of Pharmacology, School of Pharmacy , China Pharmaceutical University , Nanjing , China
| | - Chen Xue Zhuo
- a Department of Pharmacology, School of Pharmacy , China Pharmaceutical University , Nanjing , China
| | - Bashar Talal
- c Department of Pharmacy Practice, JSS College of Pharmacy , JSS University , Mysuru , India
| | - Abdoh Taleb
- a Department of Pharmacology, School of Pharmacy , China Pharmaceutical University , Nanjing , China
| | - Enos Mais
- d Department of Pharmacognosy, School of Pharmacy , China Pharmaceutical University , Nanjing , China
| | - Ding Qilong
- a Department of Pharmacology, School of Pharmacy , China Pharmaceutical University , Nanjing , China
| |
Collapse
|
160
|
Endothelial Alterations in Systemic Lupus Erythematosus and Rheumatoid Arthritis: Potential Effect of Monocyte Interaction. Mediators Inflamm 2017; 2017:9680729. [PMID: 28546658 PMCID: PMC5435976 DOI: 10.1155/2017/9680729] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 12/20/2022] Open
Abstract
Patients with systemic autoimmune diseases such as rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) are prone to develop atherosclerosis and cardiovascular diseases five times more often than the general population; this increase in frequency could be partially explained by an increase in the macrovasculature endothelial damage. In these autoimmune diseases, a microvascular endothelial injury has also been reported in different organs and tissues, especially in sites where ultrafiltration processes occur. Different components that are characteristic to the immunopathology of RA and SLE could be involved in the endothelial cell activation, permeability increase, functional alteration, and vascular injury. Circulating immune complexes (IC) detected in SLE and RA have been proposed to participate in the endothelial injury. In the vascular environment, IC can generate different responses that could be mediated by monocytes, because these cells have patrolling and monitoring functions on the endothelium. However, with certain stimuli such as TLR ligands, the monocytes are retained in the lumen, releasing proinflammatory mediators that participate in the endothelial damage. This paper aims to review some aspects about the endothelial activation and dysfunction in the context of SLE and RA, as well as the potential role that monocytes apparently play in this process.
Collapse
|
161
|
Montanari A, Lazzeroni D, Pelà G, Crocamo A, Lytvyn Y, Musiari L, Cabassi A, Cherney DZI. Calcium channel blockade blunts the renal effects of acute nitric oxide synthase inhibition in healthy humans. Am J Physiol Renal Physiol 2017; 312:F870-F878. [DOI: 10.1152/ajprenal.00568.2016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 02/01/2017] [Accepted: 02/06/2017] [Indexed: 01/25/2023] Open
Abstract
Our aim was to investigate whether blockade of calcium channels (CCs) or angiotensin II type 1 receptors (AT1R) modulates renal responses to nitric oxide synthesis inhibition (NOSI) in humans. Fourteen sodium-replete, healthy volunteers underwent 90-min infusions of 3.0 μg·kg−1·min−1 NG-nitro-l-arginine methyl ester (l-NAME) on 3 occasions, preceded by 3 days of either placebo (PL), 10 mg of manidipine (MANI), or 50 mg of losartan (LOS). At each phase, mean arterial pressure (MAP), glomerular filtration rate (GFR; inulin), renal blood flow (RBF; p-aminohippurate), urinary sodium (UNaV), and 8-isoprostane (U8-iso-PGF2αV; an oxidative stress marker) were measured. With PL + l -NAME, the following changes were observed: +6% MAP ( P < 0.005 vs. baseline), −10% GFR, −20% RBF, −49% UNaV ( P < 0.001), and +120% U8-iso-PGF2αV ( P < 0.01). In contrast, MAP did not increase during LOS + l-NAME or MANI + l-NAME ( P > 0.05 vs. baseline), whereas renal changes were the same during LOS + l-NAME vs. PL + l-NAME (ANOVA, P > 0.05). However, during MANI + l-NAME, changes vs. baseline in GFR (−6%), RBF (−12%), and UNaV (−34%) were blunted vs. PL + l-NAME and LOS + l-NAME ( P < 0.005), and the rise in U8-iso-PGF2αV was almost abolished (+37%, P > 0.05 vs. baseline; P < 0.01 vs. PL + l-NAME or LOS + l-NAME). We conclude that, since MANI blunted l-NAME-induced renal hemodynamic changes, CCs participate in the renal responses to NOSI in healthy, sodium-replete humans independent of changes in MAP and without the apparent contribution of the AT1R. Because the rise in U8-iso-PGF2αV was essentially prevented during MANI + l-NAME, CC blockade may oppose the renal effects of NOSI in part by counteracting oxidative stress responses to acutely impaired renal NO bioavailability.
Collapse
Affiliation(s)
- Alberto Montanari
- Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy
| | - Davide Lazzeroni
- Prevention and Rehabilitation Unit at the Don Gnocchi Foundation and Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy; and
| | - Giovanna Pelà
- Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy
| | - Antonio Crocamo
- Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy
| | - Yuliya Lytvyn
- Division of Nephrology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Luisa Musiari
- Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy
| | - Aderville Cabassi
- Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy
| | - David Z. I. Cherney
- Division of Nephrology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
162
|
Silva GC, Abbas M, Khemais-Benkhiat S, Burban M, Ribeiro TP, Toti F, Idris-Khodja N, Côrtes SF, Schini-Kerth VB. Replicative senescence promotes prothrombotic responses in endothelial cells: Role of NADPH oxidase- and cyclooxygenase-derived oxidative stress. Exp Gerontol 2017; 93:7-15. [PMID: 28412252 DOI: 10.1016/j.exger.2017.04.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 03/01/2017] [Accepted: 04/11/2017] [Indexed: 12/15/2022]
Abstract
Endothelial senescence has been suggested to promote endothelial dysfunction in age-related vascular disorders. This study evaluated the prothrombotic properties of senescent endothelial cells (ECs) and the underlying mechanism. Serial passaging from passage (P)1 to P4 (replicative senescence) of porcine coronary artery ECs, or treatment of P1 ECs with the endothelial nitric oxide synthase (eNOS) inhibitor L-NAME (premature senescence) induced acquisition of markers of senescence including increased senescence-associated-β-galactosidase (SA-β-gal) activity and p53, p21, p16 expression. Approximately 55% of P3 cells were senescent with a high level oxidative stress, and decreased eNOS-derived nitric oxide (NO) formation associated with increased expression of NADPH oxidase subunits (gp91phox, p47phox), cyclooxygenase (COX)-2 but not COX-1, and a decreased eNOS expression leading to a reduced ability of ECs to inhibit platelet aggregation. P3 cells also presented increased expression and activity of tissue factor (TF), a key initiator of the coagulation cascade. Treatment of senesecent cells with a NADPH oxidase inhibitor (VAS-2870) or by a COX inhibitor (indomethacin) reduced oxidative stress, decreased TF activity and expression, and reduced the expression of gp91phox, p47phox and COX-2 and restored the ability of ECs to inhibit effectively platelet aggregation. Thus, replicative endothelial senescence promotes a prothrombotic response involving the down-regulation of the protective NO pathway and the upregulation of the NADPH oxidase- and COXs-dependent oxidative stress pathway promoting TF expression and activity.
Collapse
Affiliation(s)
- Grazielle Caroline Silva
- Laboratoire de Biophotonique et Pharmacologie, UMR CNRS 7213, Faculté de Pharmacie, Université de Strasbourg, France; Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Malak Abbas
- Laboratoire de Biophotonique et Pharmacologie, UMR CNRS 7213, Faculté de Pharmacie, Université de Strasbourg, France
| | - Sonia Khemais-Benkhiat
- Laboratoire de Biophotonique et Pharmacologie, UMR CNRS 7213, Faculté de Pharmacie, Université de Strasbourg, France
| | - Melanie Burban
- Laboratoire de Biophotonique et Pharmacologie, UMR CNRS 7213, Faculté de Pharmacie, Université de Strasbourg, France
| | - Thais Porto Ribeiro
- Laboratoire de Biophotonique et Pharmacologie, UMR CNRS 7213, Faculté de Pharmacie, Université de Strasbourg, France
| | - Florence Toti
- Laboratoire de Biophotonique et Pharmacologie, UMR CNRS 7213, Faculté de Pharmacie, Université de Strasbourg, France
| | - Noureddine Idris-Khodja
- Laboratoire de Biophotonique et Pharmacologie, UMR CNRS 7213, Faculté de Pharmacie, Université de Strasbourg, France
| | - Steyner F Côrtes
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Valérie B Schini-Kerth
- Laboratoire de Biophotonique et Pharmacologie, UMR CNRS 7213, Faculté de Pharmacie, Université de Strasbourg, France.
| |
Collapse
|
163
|
Parallel comparison of risk factors between progression of organic stenosis in the coronary arteries and onset of acute coronary syndrome by covariance structure analysis. PLoS One 2017; 12:e0173898. [PMID: 28301565 PMCID: PMC5354387 DOI: 10.1371/journal.pone.0173898] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 02/28/2017] [Indexed: 12/14/2022] Open
Abstract
Background It is widely accepted that progression of organic stenosis in the coronary arteries and onset of acute coronary syndrome (ACS) are similar in the development of atherosclerosis. However, the extent of the association of each risk factor with the respective pathological conditions has not been fully elucidated. Objectives We investigated the differences in risk factors between these conditions using a statistical procedure. Methods The study population consisted of 1,029 patients with ischemic heart disease (IHD). We divided the study population into two groups (ACS and non-ACS) and by diseased vessels (organic stenosis). Covariance structure analysis was simultaneously performed in one equation model for determination and comparison of the risk factors for organic stenosis and ACS. Results The analysis revealed that age (standardized regression coefficient, β: 0.206, P < 0.001), male gender (β: 0.126, P < 0.001), HbA1c level (β: 0.109, P < 0.001), HDL level (β: -0.109, P < 0.001) and LDL level (β: 0.127, P = 0.002) were significant for the advancement of organic stenosis. HDL level (β: 0100, P = 0.002) and MDA-LDL level (β: 0.335, P < 0.001) were significant for the onset of ACS, but age, HbA1c and LDL (P = NS or β < 0.1, respectively) were not. Among the risk factors, age, HbA1c and LDL were significantly more strongly associated with organic stenosis than ACS, while MDA-LDL was significantly more strongly associated with ACS than organic stenosis. Conclusions The current statistical analysis revealed clear differences among the risk factors between the progression of organic stenosis and the onset of ACS. Among them, the MDA-LDL level should be considered to indicate a substantial risk of ACS.
Collapse
|
164
|
Abstract
The heart is uniquely responsible for providing its own blood supply through the coronary circulation. Regulation of coronary blood flow is quite complex and, after over 100 years of dedicated research, is understood to be dictated through multiple mechanisms that include extravascular compressive forces (tissue pressure), coronary perfusion pressure, myogenic, local metabolic, endothelial as well as neural and hormonal influences. While each of these determinants can have profound influence over myocardial perfusion, largely through effects on end-effector ion channels, these mechanisms collectively modulate coronary vascular resistance and act to ensure that the myocardial requirements for oxygen and substrates are adequately provided by the coronary circulation. The purpose of this series of Comprehensive Physiology is to highlight current knowledge regarding the physiologic regulation of coronary blood flow, with emphasis on functional anatomy and the interplay between the physical and biological determinants of myocardial oxygen delivery. © 2017 American Physiological Society. Compr Physiol 7:321-382, 2017.
Collapse
Affiliation(s)
- Adam G Goodwill
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
| | - Gregory M Dick
- California Medical Innovations Institute, 872 Towne Center Drive, Pomona, CA
| | - Alexander M Kiel
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
- Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Drive, Lafayette, IN
| | - Johnathan D Tune
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
165
|
Félétou M. Discovery of Nitric Oxide and Translation to Clinical Application. Physiology (Bethesda) 2017; 31:76-7. [PMID: 26865574 DOI: 10.1152/physiol.00056.2015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|
166
|
Battson ML, Lee DM, Gentile CL. Endoplasmic reticulum stress and the development of endothelial dysfunction. Am J Physiol Heart Circ Physiol 2017; 312:H355-H367. [DOI: 10.1152/ajpheart.00437.2016] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 11/28/2016] [Accepted: 11/28/2016] [Indexed: 12/14/2022]
Abstract
The vascular endothelium plays a critical role in cardiovascular homeostasis, and thus identifying the underlying causes of endothelial dysfunction has important clinical implications. In this regard, the endoplasmic reticulum (ER) has recently emerged as an important regulator of metabolic processes. Dysfunction within the ER, broadly termed ER stress, evokes the unfolded protein response (UPR), an adaptive pathway that aims to restore ER homeostasis. Although the UPR is the first line of defense against ER stress, chronic activation of the UPR leads to cell dysfunction and death and has recently been implicated in the pathogenesis of endothelial dysfunction. Numerous risk factors for endothelial dysfunction can induce ER stress, which may in turn disrupt endothelial function via direct effects on endothelium-derived vasoactive substances or by activating other pathogenic cellular networks such as inflammation and oxidative stress. This review summarizes the available data linking ER stress to endothelial dysfunction.
Collapse
Affiliation(s)
- M. L. Battson
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, Colorado
| | - D. M. Lee
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, Colorado
| | - C. L. Gentile
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
167
|
Ma Y, Li L, Shao Y, Bai X, Bai T, Huang X. Methotrexate improves perivascular adipose tissue/endothelial dysfunction via activation of AMPK/eNOS pathway. Mol Med Rep 2017; 15:2353-2359. [PMID: 28259947 DOI: 10.3892/mmr.2017.6225] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 11/29/2016] [Indexed: 11/06/2022] Open
Abstract
Adipose and endothelial dysfunction is associated with cardiovascular disease. Perivascular adipose tissue (PVAT) directly surrounds vessels and influences vessel function via a paracrine effect, and adenosine monophosphate (AMP)-activated protein kinase (AMPK) modulates the metabolic pathway, thus, the present study hypothesized that activation of AMPK in PVAT may regulate endothelial function in pathological settings. The present study investigated the effect of methotrexate (MTX) on adipocytokine expression in PVAT with an emphasis on the regulation of endothelial function. The effects of MTX and the mechanisms involved were investigated using a relaxation assay and western blot analysis. Reverse transcription‑quantitative polymerase chain reaction and western blotting were used to detect the mRNA and protein expression levels. ELISA assay was used to quantify the level of TNF‑α and IL‑6. Palmitic acid (PA) stimulation induced inflammation and dysregulation of adipocytokine expression in PVAT. MTX treatment inhibited nuclear factor‑κB p65 phosphorylation and downregulated expression of pro‑inflammatory cytokines, including tumor necrosis factor‑α and interleukin-6, whereas adiponectin expression increased. MTX increased AMPK phosphorylation under basal and inflammatory conditions in PVAT, whereas knockdown of AMPK via small interfering RNA diminished its modulatory effect, indicating that MTX inhibits inflammation in an AMPK‑dependent manner. The present study prepared conditioned medium from PA‑stimulated PVAT to induce endothelial dysfunction and observed that pre‑treatment of PVAT with MTX effectively restored the loss of acetylcholine‑induced vasodilation and increased endothelial nitric oxide synthase phosphorylation in the rat aorta. The results of the present study demonstrated that MTX ameliorated inflammation-associated adipocytokine dysregulation and thus prevented endothelial dysfunction. These data provide further pharmacological evidence regarding the beneficial effects of MTX in cardiovascular diseases.
Collapse
Affiliation(s)
- Yanmin Ma
- Department of Pharmacy, Ninth Hospital of Xi'an, Xi'an, Shaanxi 710054, P.R. China
| | - Li Li
- Department of Pharmacy, Ninth Hospital of Xi'an, Xi'an, Shaanxi 710054, P.R. China
| | - Yating Shao
- Department of Gerontology, Ninth Hospital of Xi'an, Xi'an, Shaanxi 710054, P.R. China
| | - Xiaohong Bai
- Department of Pharmacy, Chang'an District Hospital of Xi'an, Xi'an, Shaanxi 710018, P.R. China
| | - Tiao Bai
- Department of Gerontology, Ninth Hospital of Xi'an, Xi'an, Shaanxi 710054, P.R. China
| | - Xinliang Huang
- Department of Gerontology, Ninth Hospital of Xi'an, Xi'an, Shaanxi 710054, P.R. China
| |
Collapse
|
168
|
Abstract
Exposure to air pollution negatively impacts cardiovascular health. Studies show that increased exposure to a number of airborne pollutants increases the risk for cardiovascular disease progression, myocardial events, and cardiovascular mortality. A hypothesized mechanism linking air pollution and cardiovascular disease is the development of systemic inflammation and endothelium dysfunction, the latter of which can result from an imbalance of vasoactive factors within the vasculature. Endothelin-1 (ET-1) is a potent peptide vasoconstrictor that plays a significant role in regulating vascular homeostasis. It has been reported that the production and function of ET-1 and its receptors are upregulated in a number of disease states associated with endothelium dysfunction including hypertension and atherosclerosis. This mini-review surveys epidemiological and experimental air pollution studies focused on ET-1 dysregulation as a plausible mechanism underlying the development of cardiovascular disease. Although alterations in ET-1 system components are observed in some studies, there remains a need for future research to clarify whether these specific changes are compensatory or causally related to vascular injury and dysfunction. Moreover, further research may test the efficacy of selective ET-1 pharmacological interventions (e.g., ETA receptor inhibitors) to determine whether these treatments could impede the deleterious impact of air pollution exposure on cardiovascular health.
Collapse
|
169
|
Effects of aerobic exercise training on metabolism of nitric oxide and endothelin-1 in lung parenchyma of rats with pulmonary arterial hypertension. Mol Cell Biochem 2017; 429:73-89. [DOI: 10.1007/s11010-016-2937-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/23/2016] [Indexed: 12/19/2022]
|
170
|
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
|
171
|
Groschner K, Shrestha N, Fameli N. Cardiovascular and Hemostatic Disorders: SOCE in Cardiovascular Cells: Emerging Targets for Therapeutic Intervention. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:473-503. [PMID: 28900929 DOI: 10.1007/978-3-319-57732-6_24] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The discovery of the store-operated Ca2+ entry (SOCE) phenomenon is tightly associated with its recognition as a pathway of high (patho)physiological significance in the cardiovascular system. Early on, SOCE has been investigated primarily in non-excitable cell types, and the vascular endothelium received particular attention, while a role of SOCE in excitable cells, specifically cardiac myocytes and pacemakers, was initially ignored and remains largely enigmatic even to date. With the recent gain in knowledge on the molecular components of SOCE as well as their cellular organization within nanodomains, potential tissue/cell type-dependent heterogeneity of the SOCE machinery along with high specificity of linkage to downstream signaling pathways emerged for cardiovascular cells. The basis of precise decoding of cellular Ca2+ signals was recently uncovered to involve correct spatiotemporal organization of signaling components, and even minor disturbances in these assemblies trigger cardiovascular pathologies. With this chapter, we wish to provide an overview on current concepts of cellular organization of SOCE signaling complexes in cardiovascular cells with particular focus on the spatiotemporal aspects of coupling to downstream signaling and the potential disturbance of these mechanisms by pathogenic factors. The significance of these mechanistic concepts for the development of novel therapeutic strategies will be discussed.
Collapse
Affiliation(s)
- Klaus Groschner
- Institute of Biophysics, Medical University of Graz, Neue Stiftingtalstrasse 6/4, 8010, Graz, Austria.
| | - Niroj Shrestha
- Institute of Biophysics, Medical University of Graz, Neue Stiftingtalstrasse 6/4, 8010, Graz, Austria
| | - Nicola Fameli
- Institute of Biophysics, Medical University of Graz, Neue Stiftingtalstrasse 6/4, 8010, Graz, Austria
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
172
|
Mauricio MD, Serna E, Fernández-Murga ML, Portero J, Aldasoro M, Valles SL, Sanz Y, Vila JM. Bifidobacterium pseudocatenulatum CECT 7765 supplementation restores altered vascular function in an experimental model of obese mice. Int J Med Sci 2017; 14:444-451. [PMID: 28539820 PMCID: PMC5441036 DOI: 10.7150/ijms.18354] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 01/30/2017] [Indexed: 01/01/2023] Open
Abstract
Aims.Bifidobacterium pseudocatenulatum CECT 7765 improves metabolic and immunological altered functions in high fat fed mice, however little is known about the effects of potential probiotics on vascular reactivity. The aim of the present study was to investigate the effects of a potential probiotic strain, Bifidobacterium pseudocatenulatum CECT 7765, on vascular response in obese mice. Methods. Aorta samples were obtained from mice, which were divided into three groups: a control group, receiving a standard diet; an obese group, receiving a high-fat diet; and an obese group receiving high-fat diet and a daily dose of B. pseudocatenulatum CECT 7765 by oral gavage. Aortic rings were suspended in organ baths for isometric recording of tension. mRNA expression of eNOS was evaluated by real-time polymerase chain reaction. Results. Contractions induced by KCl, noradrenaline and thromboxane analogue were 33%, 30% and 45% lower respectively in aortic rings from obese mice. Bifidobacteria administration reversed this effect. eNOS inhibition increased the response to noradrenaline in the three groups with a significant lower magnitude in aortic rings from obese mice receiving bifidobacteria supplement. Acetylcholine caused a greater vasodilation in aorta from obese group (46±3% for control and 69±4% for obese group; p<0.05) and bifidobacteria reversed it (57±5%). Response to sodium nitroprusside was displaced 2.9 times to the left in a parallel manner in obese group. Relaxation to sodium nitroprusside remained unchanged in the bifidobacteria fed group. There was about five-fold decreased mRNA expression of eNOS in aortic segments from the group receiving bifidobacteria. Conclusion.Bifidobacterium pseudocatenulatum CECT 7765 restores the obesity-induced altered vascular function mainly by reducing nitric oxide release.
Collapse
Affiliation(s)
- María D Mauricio
- Departamento de Fisiología, Universitat de Valencia, Valencia, Spain.,Fundación de Investigación del Hospital Clínico Universitario de Valencia/INCLIVA, Valencia, Spain
| | - Eva Serna
- Unidad Central de Investigación. Facultad de Medicina, Universitat de Valencia, Valencia, Spain
| | - María Leonor Fernández-Murga
- Microbial Ecology, Nutrition and Health Research Group, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Jesica Portero
- Unidad Central de Investigación. Facultad de Medicina, Universitat de Valencia, Valencia, Spain
| | - Martín Aldasoro
- Departamento de Fisiología, Universitat de Valencia, Valencia, Spain.,Fundación de Investigación del Hospital Clínico Universitario de Valencia/INCLIVA, Valencia, Spain
| | - Soraya L Valles
- Departamento de Fisiología, Universitat de Valencia, Valencia, Spain.,Fundación de Investigación del Hospital Clínico Universitario de Valencia/INCLIVA, Valencia, Spain
| | - Yolanda Sanz
- Microbial Ecology, Nutrition and Health Research Group, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - José M Vila
- Departamento de Fisiología, Universitat de Valencia, Valencia, Spain.,Fundación de Investigación del Hospital Clínico Universitario de Valencia/INCLIVA, Valencia, Spain
| |
Collapse
|
173
|
A novel role for small molecule glycomimetics in the protection against lipid-induced endothelial dysfunction: Involvement of Akt/eNOS and Nrf2/ARE signaling. Biochim Biophys Acta Gen Subj 2017; 1861:3311-3322. [DOI: 10.1016/j.bbagen.2016.08.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/20/2016] [Accepted: 08/19/2016] [Indexed: 02/07/2023]
|
174
|
Schinzari F, Tesauro M, Cardillo C. Vascular hyperpolarization in human physiology and cardiovascular risk conditions and disease. Acta Physiol (Oxf) 2017; 219:124-137. [PMID: 28009486 DOI: 10.1111/apha.12630] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 10/05/2015] [Accepted: 11/05/2015] [Indexed: 12/11/2022]
Abstract
Hyperpolarization causing smooth muscle relaxation contributes to the maintenance of vascular homeostasis, particularly in small-calibre arteries and arterioles. It may also become a compensatory vasodilator mechanism upregulated in states with impaired nitric oxide (NO) availability. Bioassay of vascular hyperpolarization in the human circulation has been hampered by the complexity of mechanisms involved and the limited availability of investigational tools. Firm evidence, however, supports the notion that hyperpolarization participates in the regulation of resting vasodilator tone and vascular reactivity in healthy subjects. In addition, an enhanced endothelium-derived hyperpolarization contributes to both resting and agonist-stimulated vasodilation in a variety of cardiovascular risk conditions and disease. Thus, hyperpolarization mediated by epoxyeicosatrienoic acids (EETs) and H2 O2 has been observed in coronary arterioles of patients with coronary artery disease. Similarly, ouabain-sensitive and EETs-mediated hyperpolarization has been observed to compensate for NO deficiency in patients with essential hypertension. Moreover, in non-hypertensive patients with multiple cardiovascular risk factors and in hypercholesterolaemia, KCa channel-mediated vasodilation appears to be activated. A novel paradigm establishes that perivascular adipose tissue (PVAT) is an additional regulator of vascular tone/function and endothelium is not the only agent in vascular hyperpolarization. Indeed, some PVAT-derived relaxing substances, such as adiponectin and angiotensin 1-7, may exert anticontractile and vasodilator actions by the opening of KCa channels in smooth muscle cells. Conversely, PVAT-derived factors impair coronary vasodilation via differential inhibition of some K+ channels. In view of adipose tissue abnormalities occurring in human obesity, changes in PVAT-dependent hyperpolarization may be relevant for vascular dysfunction also in this condition.
Collapse
Affiliation(s)
- F. Schinzari
- Department of Internal Medicine; Catholic University; Rome Italy
| | - M. Tesauro
- Department of Internal Medicine; Tor Vergata University; Rome Italy
| | - C. Cardillo
- Department of Internal Medicine; Catholic University; Rome Italy
| |
Collapse
|
175
|
Ghebre YT, Yakubov E, Wong WT, Krishnamurthy P, Sayed N, Sikora AG, Bonnen MD. Vascular Aging: Implications for Cardiovascular Disease and Therapy. TRANSLATIONAL MEDICINE (SUNNYVALE, CALIF.) 2016; 6:183. [PMID: 28932625 PMCID: PMC5602592 DOI: 10.4172/2161-1025.1000183] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The incidence and prevalence of cardiovascular disease is highest among the elderly, in part, due to deleterious effects of advancing age on the heart and blood vessels. Aging, a known cardiovascular risk factor, is progressively associated with structural and functional changes to the vasculature including hemodynamic disturbance due to increased oxidative stress, premature cellular senescence and impairments in synthesis and/or secretion of endothelium-derived vasoactive molecules. These molecular and physiological changes lead to vessel wall stiffening and thickening, as well as other vascular complications that culminate to loss of vascular tone regulation and endothelial function. Intriguingly, the vessel wall, a biochemically active structure composed of collagen, connective tissue, smooth muscle and endothelial cells, is adversely affected by processes involved in premature or normal aging. Notably, the inner most layer of the vessel wall, the endothelium, becomes senescent and dysfunctional with advancing age. As a result, its ability to release vasoactive molecules such as acetylcholine (ACh), prostacyclin (PGI2), endothelium-derived hyperpolarizing factor (EDHF), and nitric oxide (NO) is reduced and the cellular response to these molecules is also impaired. By contrast, the vascular endothelium increases its generation and release of reactive oxygen (ROS) and nitrogen (RNS) species, vasoconstrictors such as endothelin (ET) and angiotensin (AT), and endogenous inhibitors of NO synthases (NOSs) to block NO. This skews the balance of the endothelium in favor of the release of highly tissue reactive and harmful molecules that promote DNA damage, telomere erosion, senescence, as well as stiffened and hardened vessel wall that is prone to the development of hypertension, diabetes, atherosclerosis and other cardiovascular risk factors. This Review discusses the impact of advancing age on cardiovascular health, and highlights the cellular and molecular mechanisms that underlie age-associated vascular changes. In addition, the role of pharmacological interventions in preventing or delaying age-related cardiovascular disease is discussed.
Collapse
Affiliation(s)
- Yohannes T Ghebre
- Department of Radiation Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Eduard Yakubov
- phaRNA Comprehensive RNA Technologies, Houston, Texas, USA
| | - Wing Tak Wong
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nazish Sayed
- Department of Medicine, Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Andrew G Sikora
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Mark D Bonnen
- Department of Radiation Oncology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
176
|
Zhang Y, Guan L, Yu J, Zhao Z, Mao L, Li S, Zhao J. Pulmonary endothelial activation caused by extracellular histones contributes to neutrophil activation in acute respiratory distress syndrome. Respir Res 2016; 17:155. [PMID: 27871277 PMCID: PMC5117496 DOI: 10.1186/s12931-016-0472-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/08/2016] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND During the acute respiratory distress syndrome (ARDS), neutrophils play a central role in the pathogenesis, and their activation requires interaction with the endothelium. Extracellular histones have been recognized as pivotal inflammatory mediators. This study was to investigate the role of pulmonary endothelial activation during the extracellular histone-induced inflammatory response in ARDS. METHODS ARDS was induced in male C57BL/6 mice by intravenous injection with lipopolysaccharide (LPS) or exogenous histones. Concurrent with LPS administration, anti-histone H4 antibody (anti-H4) or non-specific IgG was administered to study the role of extracellular histones. The circulating von Willebrand factor (vWF) and soluble thrombomodulin (sTM) were measured with ELISA kits at the preset time points. Myeloperoxidase (MPO) activity in lung tissue was measured with a MPO detection kit. The translocation of P-selectin and neutrophil infiltration were measured by immunohistochemical detection. For in vitro studies, histone H4 in the supernatant of mouse lung vascular endothelial cells (MLVECs) was measured by Western blot. The binding of extracellular histones with endothelial membrane was examined by confocal laser microscopy. Endothelial P-selectin translocation was measured by cell surface ELISA. Adhesion of neutrophils to MLVECs was assessed with a color video digital camera. RESULTS The results showed that during LPS-induced ARDS extracellular histones caused endothelial and neutrophil activation, as seen by P-selectin translocation, release of vWF, an increase of circulating sTM, lung neutrophil infiltration and increased MPO activity. Extracellular histones directly bound and activated MLVECs in a dose-dependent manner. On the contrary, the direct stimulatory effect of exogenous histones on neutrophils was very limited, as measured by neutrophil adhesion and MPO activity. With the contribution of activated endothelium, extracellular histones could effectively activating neutrophils. Both inhibiting the endothelial activation with an anti-toll like receptor (TLR) antibody and inhibiting the interaction of the endothelium with neutrophil using an anti-P-selectin antibody decreased the degree of neutrophil activation. CONCLUSIONS Extracellular histones are pro-inflammatory mediators in LPS-induced ARDS in mice. In addition to direct action to neutrophils, extracellular histones promote neutrophil adhesion and subsequent activation by first activating the pulmonary endothelium via TLR signaling. Thus, endothelial activation is important for extracellular histone-induced inflammatory injury.
Collapse
Affiliation(s)
- Yanlin Zhang
- Research Center of Occupational Medicine, Third Hospital of Peking University, No.49 North Garden Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Li Guan
- Research Center of Occupational Medicine, Third Hospital of Peking University, No.49 North Garden Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Jie Yu
- Research Center of Occupational Medicine, Third Hospital of Peking University, No.49 North Garden Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Zanmei Zhao
- Research Center of Occupational Medicine, Third Hospital of Peking University, No.49 North Garden Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Lijun Mao
- Research Center of Occupational Medicine, Third Hospital of Peking University, No.49 North Garden Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Shuqiang Li
- Research Center of Occupational Medicine, Third Hospital of Peking University, No.49 North Garden Road, Haidian District, Beijing, 100191, People's Republic of China.
| | - Jinyuan Zhao
- Research Center of Occupational Medicine, Third Hospital of Peking University, No.49 North Garden Road, Haidian District, Beijing, 100191, People's Republic of China.
| |
Collapse
|
177
|
Hayat U, Thondapu V, Tsay T, Barlis P. Atherogenesis and Inflammation. Interv Cardiol 2016. [DOI: 10.1002/9781118983652.ch1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Umair Hayat
- Melbourne Medical School; The University of Melbourne; Australia
| | - Vikas Thondapu
- Melbourne Medical School; The University of Melbourne; Australia
| | - Tim Tsay
- Melbourne Medical School; The University of Melbourne; Australia
| | | |
Collapse
|
178
|
Roos CM, Zhang B, Palmer AK, Ogrodnik MB, Pirtskhalava T, Thalji NM, Hagler M, Jurk D, Smith LA, Casaclang‐Verzosa G, Zhu Y, Schafer MJ, Tchkonia T, Kirkland JL, Miller JD. Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell 2016; 15:973-7. [PMID: 26864908 PMCID: PMC5013022 DOI: 10.1111/acel.12458] [Citation(s) in RCA: 515] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2016] [Indexed: 12/19/2022] Open
Abstract
While reports suggest a single dose of senolytics may improve vasomotor function, the structural and functional impact of long‐term senolytic treatment is unknown. To determine whether long‐term senolytic treatment improves vasomotor function, vascular stiffness, and intimal plaque size and composition in aged or hypercholesterolemic mice with established disease. Senolytic treatment (intermittent treatment with Dasatinib + Quercetin via oral gavage) resulted in significant reductions in senescent cell markers (TAF+ cells) in the medial layer of aorta from aged and hypercholesterolemic mice, but not in intimal atherosclerotic plaques. While senolytic treatment significantly improved vasomotor function (isolated organ chamber baths) in both groups of mice, this was due to increases in nitric oxide bioavailability in aged mice and increases in sensitivity to NO donors in hypercholesterolemic mice. Genetic clearance of senescent cells in aged normocholesterolemic INK‐ATTAC mice phenocopied changes elicited by D+Q. Senolytics tended to reduce aortic calcification (alizarin red) and osteogenic signaling (qRT–PCR, immunohistochemistry) in aged mice, but both were significantly reduced by senolytic treatment in hypercholesterolemic mice. Intimal plaque fibrosis (picrosirius red) was not changed appreciably by chronic senolytic treatment. This is the first study to demonstrate that chronic clearance of senescent cells improves established vascular phenotypes associated with aging and chronic hypercholesterolemia, and may be a viable therapeutic intervention to reduce morbidity and mortality from cardiovascular diseases.
Collapse
Affiliation(s)
| | - Bin Zhang
- Department of Surgery Mayo Clinic Rochester MN USA
| | | | - Mikolaj B. Ogrodnik
- Kogod Center on Aging Mayo Clinic Rochester MN USA
- Newcastle University Institute for Aging Newcastle University Newcastle Upon Tyne UK
| | | | | | | | - Diana Jurk
- Newcastle University Institute for Aging Newcastle University Newcastle Upon Tyne UK
| | | | | | - Yi Zhu
- Kogod Center on Aging Mayo Clinic Rochester MN USA
| | | | | | - James L. Kirkland
- Kogod Center on Aging Mayo Clinic Rochester MN USA
- Department of Physiology & Biomedical Engineering Mayo Clinic Rochester MN USA
| | - Jordan D. Miller
- Department of Surgery Mayo Clinic Rochester MN USA
- Kogod Center on Aging Mayo Clinic Rochester MN USA
- Department of Physiology & Biomedical Engineering Mayo Clinic Rochester MN USA
| |
Collapse
|
179
|
Dolinina J, Sverrisson K, Rippe A, Öberg CM, Rippe B. Nitric oxide synthase inhibition causes acute increases in glomerular permeability in vivo, dependent upon reactive oxygen species. Am J Physiol Renal Physiol 2016; 311:F984-F990. [PMID: 27681559 DOI: 10.1152/ajprenal.00152.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 09/21/2016] [Indexed: 11/22/2022] Open
Abstract
There is increasing evidence that the permeability of the glomerular filtration barrier (GFB) is partly regulated by a balance between the bioavailability of nitric oxide (NO) and that of reactive oxygen species (ROS). It has been postulated that normal or moderately elevated NO levels protect the GFB from permeability increases, whereas ROS, through reducing the bioavailability of NO, have the opposite effect. We tested the tentative antagonism between NO and ROS on glomerular permeability in anaesthetized Wistar rats, in which the left ureter was cannulated for urine collection while simultaneously blood access was achieved. Rats were systemically infused with either l-NAME or l-NAME together with the superoxide scavenger Tempol, or together with l-arginine or the NO-donor DEA-NONOate, or the cGMP agonist 8-bromo-cGMP. To measure glomerular sieving coefficients (theta, θ) to Ficoll, rats were infused with FITC-Ficoll 70/400 (mol/radius 10-80 Å). Plasma and urine samples were analyzed by high-performance size-exclusion chromatography (HPSEC) for determination of θ for Ficoll repeatedly during up to 2 h. l-NAME increased θ for Ficoll70Å from 2.27 ± 1.30 × 10-5 to 8.46 ± 2.06 × 10-5 (n = 6, P < 0.001) in 15 min. Tempol abrogated these increases in glomerular permeability and an inhibition was also observed with l-arginine and with 8-bromo-cGMP. In conclusion, acute NO synthase inhibition in vivo by l-NAME caused rapid increases in glomerular permeability, which could be reversed by either an ROS antagonist or by activating the guanylyl cyclase-cGMP pathway. The data strongly suggest a protective effect of NO in maintaining normal glomerular permeability in vivo.
Collapse
Affiliation(s)
| | | | - Anna Rippe
- Department of Nephrology, Lund University, Lund, Sweden
| | - Carl M Öberg
- Department of Nephrology, Lund University, Lund, Sweden
| | - Bengt Rippe
- Department of Nephrology, Lund University, Lund, Sweden
| |
Collapse
|
180
|
Leung SB, Zhang H, Lau CW, Lin ZX. Attenuation of blood pressure in spontaneously hypertensive rats by acupuncture was associated with reduction oxidative stress and improvement from endothelial dysfunction. Chin Med 2016; 11:38. [PMID: 27582785 PMCID: PMC5006281 DOI: 10.1186/s13020-016-0110-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 08/12/2016] [Indexed: 02/07/2023] Open
Abstract
Background Hypertension can be treated effectively by acupuncture; however, the association between acupuncture and endothelial function remains unknown. This study aimed to investigate the effects of acupuncture on endothelial dysfunction and oxidative stress-related parameters in spontaneously hypertensive animals. Methods Eighteen-week-old Wistar–Kyoto rats (WKYs) and spontaneously hypertensive rats (SHRs) were arbitrarily divided into four groups: WKY control (n = 8), SHR control (n = 8), SHR sham-acupuncture (n = 8) and SHR acupuncture (n = 8). The SHR acupuncture group had electroacupuncture for 6 consecutive weeks on acupoints ST36 and LR3. Blood pressure was monitored during the treatment period, and animals were euthanized at the 6th week. Aortas were harvested for determination of angiotensin II levels, NADPH oxidase activity and nitrate/nitrite levels. The level of reactive oxygen species (ROS) was determined by dihydroethidium (DHE) imaging, and functional studies were performed to assess vascular reactivity. Endothelial nitric oxide synthase was measured by Western blot assay. Results Blood pressure at the end of treatment was significantly lower in the SHR acupuncture group (185.0 ± 5.6 mmHg) compared with the SHR sham-acupuncture and the SHR control groups (201.0 ± 5.4 and 197.4 ± 5.9 mmHg, respectively; P < 0.001). Serum angiotensin II level in the SHR control group was significantly higher than in the WKY control group (P < 0.001), while it was significantly attenuated by acupuncture treatment (P = 0.023). DHE staining showed that ROS level was reduced in the aortas (P = 0.0017) and carotid arteries (P = 0.039) of acupuncture-treated SHRs. Biochemical assays showed that acupuncture inhibited the NADPH oxidase activity (P = 0.022) and enhanced antioxidant capacity (P = 0.0039). In functional studies, endothelium-dependent relaxation of aortic rings (P = 0.018) and carotid arteries (P = 0.022) in response to acetylcholine was improved in the SHR acupuncture group. Aortas of SHRs receiving acupuncture also expressed an elevated level of eNOS (P > 0.001) and p-eNOS (P = 0.012) and a reduced nitrotyrosine level (P = 0.0012). The nitrate/nitrite level in aortic tissue was also attenuated after acupuncture (P = 0.0018). Conclusion The effects of acupuncture in treating hypertension were associated with reduced oxidative stress, increased nitric oxide bioavailability and endothelial function in SHRs. Electronic supplementary material The online version of this article (doi:10.1186/s13020-016-0110-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sin Bond Leung
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, People's Republic of China
| | - Hongwei Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, People's Republic of China
| | - Chi Wai Lau
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, People's Republic of China
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, People's Republic of China
| |
Collapse
|
181
|
Matsumoto T, Watanabe S, Yamada K, Ando M, Iguchi M, Taguchi K, Kobayashi T. Relaxation Induced by Atrial Natriuretic Peptide Is Impaired in Carotid but Not Renal Arteries from Spontaneously Hypertensive Rats Due to Reduced BKCa Channel Activity. Biol Pharm Bull 2016; 38:1801-8. [PMID: 26521831 DOI: 10.1248/bpb.b15-00527] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Atrial natriuretic peptide (ANP) plays an important role in vascular functions such as blood pressure regulation and relaxant activity. Individual vascular beds exhibit differences in vascular reactivity to various ligands, however, the difference in responsiveness to ANP between carotid and renal arteries and the molecular mechanisms of its vasorelaxant activity in a pathophysiological state, including hypertension, remain unclear. We therefore investigated this issue by exposing carotid and renal artery rings obtained from spontaneously hypertensive rats (SHR) to ANP. In the SHR artery (vs. control WKY artery), the ANP-induced relaxations were reduced in carotid artery but not renal artery. Acetylcholine-induced relaxations were reduced in both arteries in SHR (vs. WKY). Sodium nitroprusside-induced relaxation was similar in both arteries between the groups. In carotid arteries, the ANP-induced relaxation was not affected by endothelial denudation or by treatment with inhibitors of nitric oxide synthase, cyclooxygenase, the voltage-dependent potassium channel, or ATP-sensitive potassium channel in arteries from both SHR and WKY. In the carotid artery from WKY but not SHR, the ANP-induced relaxation was significantly reduced by inhibition of the large-conductance calcium-activated potassium channel (BKCa). The BKCa activator-induced relaxation was reduced in the SHR artery (vs. WKY). These results suggest that ANP-induced relaxation is impaired in the carotid artery from SHR and this impairment may be at least in part due to the reduction of BKCa activity rather than endothelial components.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University
| | | | | | | | | | | | | |
Collapse
|
182
|
Gabriele M, Pucci L, La Marca M, Lucchesi D, Della Croce CM, Longo V, Lubrano V. A fermented bean flour extract downregulates LOX-1, CHOP and ICAM-1 in HMEC-1 stimulated by ox-LDL. Cell Mol Biol Lett 2016; 21:10. [PMID: 28536613 PMCID: PMC5415722 DOI: 10.1186/s11658-016-0015-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/09/2016] [Indexed: 02/07/2023] Open
Abstract
This study focused on an extract from fermented flour from the Lady Joy variety of the common bean Phaseolus vulgaris. The extract, Lady Joy lysate (Lys LJ), is enriched in antioxidant compounds during the fermentation. We assessed it for its protective effect on endothelial cells treated with oxidized-LDL (ox-LDL). The oxidative stress was determined by measuring the contents of thiobarbituric acid-reactive substances and reactive oxygen metabolites. ICAM-1, ET-1 and IL-6 concentrations were assessed using ELISA. LOX-1 and CHOP expression were analyzed using both quantitative RT-PCR and ELISA or western blotting. Ox-LDL treatment induced significant oxidative stress, which was strongly reduced by pre-treatment with the extract. The ox-LDL exposure significantly enhanced ICAM-1, IL-6 and ET-1 levels over basal levels. Lys LJ pre-treatment exerted an inhibitory effect on ox-LDL-induced endothelial activation with ICAM-1 levels comparable to those for the untreated cells. IL-6 and ET-1 production, although reduced, was still significantly higher than for the control. Both LOX-1 and CHOP expression were upregulated after ox-LDL exposure, but this effect was significantly decreased after Lys LJ pre-treatment. Lys LJ alone did not alter the ICAM-1, IL-6 and ET-1 concentrations or CHOP expression, but it did significantly lower the LOX-1 protein level. Our data suggest that Lys LJ is an effective antioxidant that is able to inhibit the oxidation process, but that it is only marginally active against inflammation and ET-1 production in HMEC-1 exposed to ox-LDL.
Collapse
Affiliation(s)
- Morena Gabriele
- National Research Council (CNR), Institute of Biology and Agricultural Biotechnology (IBBA), Pisa Unit, Research Area of Pisa, Via Moruzzi 1, 56124 Pisa, Italy
| | - Laura Pucci
- National Research Council (CNR), Institute of Biology and Agricultural Biotechnology (IBBA), Pisa Unit, Research Area of Pisa, Via Moruzzi 1, 56124 Pisa, Italy
| | - Margherita La Marca
- National Research Council (CNR), Institute of Biology and Agricultural Biotechnology (IBBA), Pisa Unit, Research Area of Pisa, Via Moruzzi 1, 56124 Pisa, Italy
| | - Daniela Lucchesi
- Department of Clinical and Experimental Medicine, Section of Metabolic Diseases, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Clara Maria Della Croce
- National Research Council (CNR), Institute of Biology and Agricultural Biotechnology (IBBA), Pisa Unit, Research Area of Pisa, Via Moruzzi 1, 56124 Pisa, Italy
| | - Vincenzo Longo
- National Research Council (CNR), Institute of Biology and Agricultural Biotechnology (IBBA), Pisa Unit, Research Area of Pisa, Via Moruzzi 1, 56124 Pisa, Italy
| | - Valter Lubrano
- Fondazione CNR/Regione Toscana G. Monasterio, Via Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
183
|
Zhang J, Fallahzadeh MK, McCullough PA. Aging Male Spontaneously Hypertensive Rat as an Animal Model for the Evaluation of the Interplay between Contrast-Induced Acute Kidney Injury and Cardiorenal Syndrome in Humans. Cardiorenal Med 2016; 7:1-10. [PMID: 27994597 DOI: 10.1159/000447542] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Although there are some animal models for biomarkers of contrast-induced acute kidney injury (CI-AKI), for cardiorenal syndrome (CRS) and for acute renal failure, the interplay between CI-AKI and CRS has yet to be evaluated. Insight into the pathogenesis of CRS is urgently needed from animal models in order to foster the discovery and implementation of novel biomarkers for this disease. Specially designed animal models for type 1 and 3 CRS, particularly CI-AKI, have not yet emerged. SUMMARY We hypothesize that the aging male spontaneously hypertensive rat (SHR) is likely to be a suitable model. The SHR model is able to mimic risk factors for preclinical CRS that appears in the clinical setting, specifically hypertension, age, preexisting damage and dysfunction of the heart and kidney, endothelial dysfunction, increased level of reactive oxygen species, decreased level and bioavailability of nitric oxide (NO), impairment of the L-arginine-NO pathway, and insulin resistance. In the SHR, CI-AKI results in a different profile of AKI biomarkers than is seen with preexisting chronic kidney injury. KEY MESSAGES The SHR model can be used to evaluate the interaction between CI-AKI and CRS type 1 and 3 and to verify neutrophil gelatinase-associated lipocalin (NGAL) as a reliable CI-AKI biomarker for clinical application. Further research is warranted with a large number of aging male SHRs to prove NGAL as a sensitive, specific, highly predictive, early biomarker for CI-AKI.
Collapse
Affiliation(s)
- Jun Zhang
- Baylor Heart and Vascular Institute, Dallas, Tex., USA
| | | | - Peter A McCullough
- Baylor Heart and Vascular Institute, Dallas, Tex., USA; Department of Internal Medicine, Baylor University Medical Center, Dallas, Tex., USA; Baylor Jack and Jane Hamilton Heart and Vascular Hospital, Dallas, Tex, Tex., USA; The Heart Hospital Baylor Plano, Plano, Tex., USA
| |
Collapse
|
184
|
Wang YNZ, Shan K, Yao MD, Yao J, Wang JJ, Li X, Liu B, Zhang YY, Ji Y, Jiang Q, Yan B. Long Noncoding RNA-GAS5: A Novel Regulator of Hypertension-Induced Vascular Remodeling. Hypertension 2016; 68:736-48. [PMID: 27432865 DOI: 10.1161/hypertensionaha.116.07259] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/17/2016] [Indexed: 12/14/2022]
Abstract
Vascular remodeling is an important pathological feature of hypertension, leading to increased vascular resistance and reduced compliance. Endothelial cell (EC) and vascular smooth muscle cell (VSMC) dysfunction is involved in vascular remodeling. Long noncoding RNAs are potential regulators of EC and VSMC function. Herein, we determined whether long noncoding RNA-growth arrest-specific 5 (GAS5) is involved in hypertension-related vascular remodeling. We revealed that GAS5 knockdown aggravated hypertension-induced microvascular dysfunction as shown by increased retinal neovascularization and capillary leakage. GAS5 regulated the remodeling of arteries, including caudal arteries, carotid arteries, renal arteries, and thoracic arteries. GAS5 was mainly expressed in ECs and VSMCs, and its expression was significantly downregulated in hypertension. GAS5 knockdown affected endothelial activation, endothelial proliferation, VSMC phenotypic conversion, and EC-VSMC communication in vivo and in vitro. Mechanistically, GAS5 regulated EC and VSMC function through β-catenin signaling. This study identified GAS5 as a critical regulator in hypertension and demonstrated the potential of gene therapy and drug development for treating hypertension.
Collapse
Affiliation(s)
- Yang-Ning-Zhi Wang
- From the Eye Hospital, Nanjing Medical University, China (Y.-N.-Z.W., K.S., M.-D.Y., J.Y., Q.J.); Department of Ophthalmology, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (J.-J.W.); Department of Cardiology, the first Affiliated Hospital, Chongqin Medical University, China (X.L.); Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (B.L.); Department of Cardiac Surgery, the first School of Clinical Medicine, Nanjing Medical University, China (Y.-Y.Z.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (Y.J.); and Research Center, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (B.Y.)
| | - Kun Shan
- From the Eye Hospital, Nanjing Medical University, China (Y.-N.-Z.W., K.S., M.-D.Y., J.Y., Q.J.); Department of Ophthalmology, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (J.-J.W.); Department of Cardiology, the first Affiliated Hospital, Chongqin Medical University, China (X.L.); Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (B.L.); Department of Cardiac Surgery, the first School of Clinical Medicine, Nanjing Medical University, China (Y.-Y.Z.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (Y.J.); and Research Center, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (B.Y.)
| | - Mu-Di Yao
- From the Eye Hospital, Nanjing Medical University, China (Y.-N.-Z.W., K.S., M.-D.Y., J.Y., Q.J.); Department of Ophthalmology, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (J.-J.W.); Department of Cardiology, the first Affiliated Hospital, Chongqin Medical University, China (X.L.); Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (B.L.); Department of Cardiac Surgery, the first School of Clinical Medicine, Nanjing Medical University, China (Y.-Y.Z.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (Y.J.); and Research Center, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (B.Y.)
| | - Jin Yao
- From the Eye Hospital, Nanjing Medical University, China (Y.-N.-Z.W., K.S., M.-D.Y., J.Y., Q.J.); Department of Ophthalmology, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (J.-J.W.); Department of Cardiology, the first Affiliated Hospital, Chongqin Medical University, China (X.L.); Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (B.L.); Department of Cardiac Surgery, the first School of Clinical Medicine, Nanjing Medical University, China (Y.-Y.Z.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (Y.J.); and Research Center, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (B.Y.)
| | - Jia-Jian Wang
- From the Eye Hospital, Nanjing Medical University, China (Y.-N.-Z.W., K.S., M.-D.Y., J.Y., Q.J.); Department of Ophthalmology, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (J.-J.W.); Department of Cardiology, the first Affiliated Hospital, Chongqin Medical University, China (X.L.); Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (B.L.); Department of Cardiac Surgery, the first School of Clinical Medicine, Nanjing Medical University, China (Y.-Y.Z.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (Y.J.); and Research Center, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (B.Y.)
| | - Xiang Li
- From the Eye Hospital, Nanjing Medical University, China (Y.-N.-Z.W., K.S., M.-D.Y., J.Y., Q.J.); Department of Ophthalmology, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (J.-J.W.); Department of Cardiology, the first Affiliated Hospital, Chongqin Medical University, China (X.L.); Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (B.L.); Department of Cardiac Surgery, the first School of Clinical Medicine, Nanjing Medical University, China (Y.-Y.Z.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (Y.J.); and Research Center, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (B.Y.)
| | - Ban Liu
- From the Eye Hospital, Nanjing Medical University, China (Y.-N.-Z.W., K.S., M.-D.Y., J.Y., Q.J.); Department of Ophthalmology, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (J.-J.W.); Department of Cardiology, the first Affiliated Hospital, Chongqin Medical University, China (X.L.); Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (B.L.); Department of Cardiac Surgery, the first School of Clinical Medicine, Nanjing Medical University, China (Y.-Y.Z.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (Y.J.); and Research Center, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (B.Y.)
| | - Yang-Yang Zhang
- From the Eye Hospital, Nanjing Medical University, China (Y.-N.-Z.W., K.S., M.-D.Y., J.Y., Q.J.); Department of Ophthalmology, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (J.-J.W.); Department of Cardiology, the first Affiliated Hospital, Chongqin Medical University, China (X.L.); Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (B.L.); Department of Cardiac Surgery, the first School of Clinical Medicine, Nanjing Medical University, China (Y.-Y.Z.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (Y.J.); and Research Center, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (B.Y.)
| | - Yong Ji
- From the Eye Hospital, Nanjing Medical University, China (Y.-N.-Z.W., K.S., M.-D.Y., J.Y., Q.J.); Department of Ophthalmology, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (J.-J.W.); Department of Cardiology, the first Affiliated Hospital, Chongqin Medical University, China (X.L.); Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (B.L.); Department of Cardiac Surgery, the first School of Clinical Medicine, Nanjing Medical University, China (Y.-Y.Z.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (Y.J.); and Research Center, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (B.Y.)
| | - Qin Jiang
- From the Eye Hospital, Nanjing Medical University, China (Y.-N.-Z.W., K.S., M.-D.Y., J.Y., Q.J.); Department of Ophthalmology, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (J.-J.W.); Department of Cardiology, the first Affiliated Hospital, Chongqin Medical University, China (X.L.); Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (B.L.); Department of Cardiac Surgery, the first School of Clinical Medicine, Nanjing Medical University, China (Y.-Y.Z.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (Y.J.); and Research Center, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (B.Y.).
| | - Biao Yan
- From the Eye Hospital, Nanjing Medical University, China (Y.-N.-Z.W., K.S., M.-D.Y., J.Y., Q.J.); Department of Ophthalmology, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (J.-J.W.); Department of Cardiology, the first Affiliated Hospital, Chongqin Medical University, China (X.L.); Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China (B.L.); Department of Cardiac Surgery, the first School of Clinical Medicine, Nanjing Medical University, China (Y.-Y.Z.); Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (Y.J.); and Research Center, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China (B.Y.).
| |
Collapse
|
185
|
D'Onofrio N, Servillo L, Giovane A, Casale R, Vitiello M, Marfella R, Paolisso G, Balestrieri ML. Ergothioneine oxidation in the protection against high-glucose induced endothelial senescence: Involvement of SIRT1 and SIRT6. Free Radic Biol Med 2016; 96:211-22. [PMID: 27101740 DOI: 10.1016/j.freeradbiomed.2016.04.013] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 04/13/2016] [Accepted: 04/15/2016] [Indexed: 12/21/2022]
Abstract
Ergothioneine (Egt), the betaine of 2-mercapto-L-histidine, is a dietary antioxidant protecting against many diseases, including cardiovascular disease (CVD), through a redox mechanism different from alkylthiols. Here, experiments were designed to evaluate the mechanisms underlying the beneficial effect of Egt against hyperglycaemia-induced senescence in endothelial cells. To this end, cells were incubated with increasing concentrations of Egt (0.01-1.00mM) for 12h followed by incubation for 48h with high-glucose (25mM). Cell evaluation indicated that viability was not affected by mM concentrations of Egt and that the high-glucose cytotoxicity was prevented with the highest efficacy at 0.5mM Egt. The cytoprotective effect of Egt was paralleled by reduced ROS production, cell senescence, and, interestingly, the formation of hercynine (EH), a betaine we recently found to be produced during the Egt oxidation pathway. Notably, the Egt beneficial effect was exerted through the upregulation of sirtuin 1 (SIRT1) and sirtuin 6 (SIRT6) expression and the downregulation of p66Shc and NF-κB. SIRT1 activity inhibition and SIRT6 gene silencing by small interfering RNA abolished the protective effect of Egt against the high-glucose-induced endothelial senescence. These data provide the first evidence of the Egt ability to interfere with endothelial senescence linked to hyperglycaemia through the regulation of SIRT1 and SIRT6 signaling, thus further strengthening the already assessed role of these two histone deacetylases in type 2 diabetes.
Collapse
Affiliation(s)
- Nunzia D'Onofrio
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Luigi Servillo
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Alfonso Giovane
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Rosario Casale
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Milena Vitiello
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Raffaele Marfella
- Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, Second University of Naples, Naples, Italy
| | - Giuseppe Paolisso
- Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, Second University of Naples, Naples, Italy
| | - Maria Luisa Balestrieri
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy.
| |
Collapse
|
186
|
Velasquez MT, Beddhu S, Nobakht E, Rahman M, Raj DS. Ambulatory Blood Pressure in Chronic Kidney Disease: Ready for Prime Time? Kidney Int Rep 2016; 1:94-104. [PMID: 28164170 PMCID: PMC5283800 DOI: 10.1016/j.ekir.2016.05.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 05/15/2016] [Accepted: 05/18/2016] [Indexed: 12/15/2022] Open
Abstract
Hypertension is common in patients with chronic kidney disease (CKD) and is the most important modifiable risk factor for CKD progression and adverse cardiovascular events in these patients. Diagnosis and successful management of hypertension are critically dependent on accurate blood pressure (BP) measurement. This is most relevant to CKD patients, in whom BP control is difficult to achieve and in whom early antihypertensive treatment is imperative to prevent kidney and cardiovascular complications. Accumulated data indicate that ambulatory blood pressure monitoring (ABPM) is better in detecting hypertension than office BP measurement. ABPM is also a superior prognostic marker compared with office BP and has successfully identified hypertensive CKD patients at increased risk. Additionally, ABPM provides information on circadian BP variation and short-term BP variability, which is associated with cardiovascular and renal outcomes. This paper reviews the evidence for the usefulness of ABPM in detection and management of hypertension in CKD patients and discusses our current understanding of the pathophysiology of altered circadian BP rhythm and variability in CKD and the role of abnormal BP patterns detected by ABPM in relation to outcomes in CKD. In addition, this Review examines the emerging role of antihypertensive chronotherapy to tailor BP management to the circadian BP pattern abnormality detected by 24-hour ABPM.
Collapse
Affiliation(s)
- Manuel T. Velasquez
- Division of Renal Diseases and Hypertension, The George Washington University, Washington, DC, USA
| | - Srinivasan Beddhu
- University of Utah School of Medicine, Kidney and Liver Clinic, Salt Lake City, Utah, USA
| | - Ehsan Nobakht
- Division of Renal Diseases and Hypertension, The George Washington University, Washington, DC, USA
| | - Mahboob Rahman
- University Hospitals Case Medical Center, Department of Medicine–Hypertension, Cleveland, Ohio, USA
| | - Dominic S. Raj
- Division of Renal Diseases and Hypertension, The George Washington University, Washington, DC, USA
| |
Collapse
|
187
|
|
188
|
Meyer MR, Barton M. Estrogens and Coronary Artery Disease: New Clinical Perspectives. ADVANCES IN PHARMACOLOGY 2016; 77:307-60. [PMID: 27451102 DOI: 10.1016/bs.apha.2016.05.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In premenopausal women, endogenous estrogens are associated with reduced prevalence of arterial hypertension, coronary artery disease, myocardial infarction, and stroke. Clinical trials conducted in the 1990s such as HERS, WHI, and WISDOM have shown that postmenopausal treatment with horse hormone mixtures (so-called conjugated equine estrogens) and synthetic progestins adversely affects female cardiovascular health. Our understanding of rapid (nongenomic) and chronic (genomic) estrogen signaling has since advanced considerably, including identification of a new G protein-coupled estrogen receptor (GPER), which like the "classical" receptors ERα and ERβ is highly abundant in the cardiovascular system. Here, we discuss the role of estrogen receptors in the pathogenesis of coronary artery disease and review natural and synthetic ligands of estrogen receptors as well as their effects in physiology, on cardiovascular risk factors, and atherosclerotic vascular disease. Data from preclinical and clinical studies using nonselective compounds activating GPER, which include selective estrogen receptor modulators such as tamoxifen or raloxifene, selective estrogen receptor downregulators such as Faslodex™ (fulvestrant/ICI 182,780), vitamin B3 (niacin), green tea catechins, and soy flavonoids such as genistein or resveratrol, strongly suggest that activation of GPER may afford therapeutic benefit for primary and secondary prevention in patients with or at risk for coronary artery disease. Evidence from preclinical studies suggest similar efficacy profiles for selective small molecule GPER agonists such as G-1 which are devoid of uterotrophic activity. Further clinical research in this area is warranted to provide opportunities for future cardiovascular drug development.
Collapse
Affiliation(s)
- M R Meyer
- Triemli City Hospital, Zürich, Switzerland.
| | - M Barton
- Molecular Internal Medicine, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
189
|
|
190
|
Fan Y, Li S, Li XL, Lin XL, Zhu CG, Xu RX, Qing P, Wu NQ, Jiang LX, Xu B, Gao RL, Li JJ. Plasma endothelin-1 level as a predictor for poor collaterals in patients with ≥95% coronary chronic occlusion. Thromb Res 2016; 142:21-25. [PMID: 27105310 DOI: 10.1016/j.thromres.2016.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 03/07/2016] [Accepted: 04/11/2016] [Indexed: 11/26/2022]
Abstract
BACKGROUND Coronary collateral circulation (CCC) plays an important role in protecting myocardium from ischemic damage. The studies on factors which impact on CCC might be of great clinical interest. The aim of the present study was to evaluate endothelin-1 (ET-1) as a potential predictor for poor or good CCC in patients with angiography-proven ≥95% coronary occlusion. METHODS We screened 1038 consecutive patients with ≥95% occlusion in at least one major epicardial coronary artery detected by coronary angiography. Of these, 663 patients were classified into the poor CCC group with Rentrop 0-2 grade collateral circulation and 375 patients into the good CCC group with Rentrop 3 grade. The association of plasma ET-1 levels with collateral status was assessed. RESULTS We found that patients in the poor CCC group had a higher ET-1 level than those in the good CCC group (0.59±0.48 vs. 0.39±0.32pmol/L, p<0.001), and the ET-1 values increased with the descent of the Rentrop grades (p for trend<0.001). Moreover, multivariate logistic regression analysis revealed an independent association between ET-1 and collateral status (odds ratio [95% CI] for poor CCC 2.27 [1.60-3.22], p<0.001). Additionally, the association presented significance in both men (odds ratio [95% CI] for poor CCC 3.18 [2.20-4.74], p<0.001) and women (odds ratio [95% CI] for poor CCC 3.10 [1.36-7.85], p=0.011) when the sex-specific analysis was performed. CONCLUSIONS Plasma ET-1 level may be a useful, easily available marker for predicting the degree of CCC in patients with ≥95% coronary chronic occlusion.
Collapse
Affiliation(s)
- Ying Fan
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Bei Li Shi Road, Xi Cheng District, Beijing, 100037, China; Department of Cardiology, The Fifth Hospital of Wuhan & Affiliated Guangci Hospital of Wuhan University, Wuhan 430050, China
| | - Sha Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Bei Li Shi Road, Xi Cheng District, Beijing, 100037, China
| | - Xiao-Lin Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Bei Li Shi Road, Xi Cheng District, Beijing, 100037, China
| | - Xian-Liang Lin
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Bei Li Shi Road, Xi Cheng District, Beijing, 100037, China
| | - Cheng-Gang Zhu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Bei Li Shi Road, Xi Cheng District, Beijing, 100037, China
| | - Rui-Xia Xu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Bei Li Shi Road, Xi Cheng District, Beijing, 100037, China
| | - Ping Qing
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Bei Li Shi Road, Xi Cheng District, Beijing, 100037, China
| | - Na-Qiong Wu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Bei Li Shi Road, Xi Cheng District, Beijing, 100037, China
| | - Li-Xin Jiang
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Bei Li Shi Road, Xi Cheng District, Beijing, 100037, China
| | - Bo Xu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Bei Li Shi Road, Xi Cheng District, Beijing, 100037, China
| | - Run-Lin Gao
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Bei Li Shi Road, Xi Cheng District, Beijing, 100037, China
| | - Jian-Jun Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167 Bei Li Shi Road, Xi Cheng District, Beijing, 100037, China.
| |
Collapse
|
191
|
Bertin R, Chen Z, Marin R, Donati M, Feltrinelli A, Montopoli M, Zambon S, Manzato E, Froldi G. Activity of myricetin and other plant-derived polyhydroxyl compounds in human LDL and human vascular endothelial cells against oxidative stress. Biomed Pharmacother 2016; 82:472-8. [PMID: 27470387 DOI: 10.1016/j.biopha.2016.05.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/11/2016] [Accepted: 05/12/2016] [Indexed: 12/22/2022] Open
Abstract
Studies indicate that oxidative modifications of endothelium and LDL play a preeminent role in atherogenesis; therefore, the preservation of the endothelial antioxidant capacity and the inhibition of LDL oxidation by use of plant-derived compounds are an appealing strategy against several vascular disorders. On this basis, baicalein, eupatorin, galangin, magnolol, myricetin, oleuropein, silibinin and bilobalide were studied against various oxidative conditions. The radical scavenging capacity was analysed using DPPH and ORAC assays. Furthermore, the LDL oxidation was detected by measuring the formation of thiobarbituric acid reactive substances (TBARS) and by monitoring the oxidation kinetics. Further, we used cultured HUVEC to investigate the activities of the polyhydroxyl compounds towards the oxidative stress induced by H2O2. The lowest levels of TBARS were observed in the presence of oleuropein and baicalein, while myricetin, magnolol and eupatorin inhibited these ones to a lesser extent. In addition, oleuropein and myricetin exhibited higher protection in copper-induced LDL oxidation kinetics. However, only myricetin and galangin showed significant protective effects against H2O2 oxidative injury in HUVEC cells. Taken all together the results indicate myricetin as the most active agent among the selected plant-derived polyhydroxyl compounds, with prominent capacities against ox-LDL and ROS production in HUVEC.
Collapse
Affiliation(s)
- Riccardo Bertin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo E. Meneghetti 2, 35131 Padova, Italy
| | - Zheng Chen
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo E. Meneghetti 2, 35131 Padova, Italy
| | - Raffaella Marin
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Maddalena Donati
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo E. Meneghetti 2, 35131 Padova, Italy
| | - Angela Feltrinelli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo E. Meneghetti 2, 35131 Padova, Italy
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo E. Meneghetti 2, 35131 Padova, Italy
| | - Sabina Zambon
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Enzo Manzato
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Guglielmina Froldi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo E. Meneghetti 2, 35131 Padova, Italy.
| |
Collapse
|
192
|
Cao Y, Fang Y, Mu J, Liu X. High salt medium activates RhoA/ROCK and downregulates eNOS expression via the upregulation of ADMA. Mol Med Rep 2016; 14:606-12. [PMID: 27175806 DOI: 10.3892/mmr.2016.5241] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 04/18/2016] [Indexed: 11/05/2022] Open
Abstract
Endothelial dysfunction has an important role in the development and progression of salt-sensitive hypertension. Asymmetric dimethylarginine (ADMA), which is an endogenous inhibitor of nitric oxide synthase (NOS), has been demonstrated to be involved in the pathophysiological processes of endothelial dysfunction and salt‑sensitive hypertension. However, it is currently unclear how high salt intake may induce these processes. The present study investigated the effects of high salt medium on ADMA, endothelial NOS (eNOS) and the Ras homolog gene family, member A (RhoA)/Rho-associated protein kinase (ROCK) pathway in the EA.hy926 umbilical vein cell line. The results demonstrated that high salt medium significantly increased the concentration of ADMA, the expression of protein arginine methyltransferase 1 (PRMT‑1) and RhoA, and the activity of ROCK, and downregulated the expression of eNOS. Knockdown of PRMT-1 with small interfering RNA (siRNA) significantly abrogated the aforementioned effects. These results indicated that ADMA has a key role in high salt‑mediated activation of the RhoA/ROCK pathway and inhibition of eNOS biosynthesis. siRNA‑PRMT‑1 may be considered a novel remedy for the treatment of endothelial dysfunction.
Collapse
Affiliation(s)
- Yu Cao
- Department of Pediatrics, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yuan Fang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jianjun Mu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaohong Liu
- Department of Pediatrics, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
193
|
|
194
|
Affiliation(s)
- Chantal M. Boulanger
- From the INSERM, U970, Paris Cardiovascular Research Center–PARCC, and Université Paris Descartes, Sorbonne Paris Cité, UMR-S970, Paris, France
| |
Collapse
|
195
|
Broseghini-Filho GB, Almenara CCP, Vassallo DV, Padilha AS. Blood Pressure Decreases Following Lead Treatment Cessation: Highest NO Bioavailability Involved. Biol Trace Elem Res 2016; 170:410-4. [PMID: 26342820 DOI: 10.1007/s12011-015-0497-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 08/28/2015] [Indexed: 10/23/2022]
Abstract
Although lead is known to induce arterial hypertension and vascular damage, it is not clear if after cessation of lead treatment, the increase of blood pressure is sustained and the vascular function is different from untreated rats. Therefore, we aimed to evaluate the systolic blood pressure during and following lead-treatment discontinuance and the possible vascular alterations involved with it. Rats received lead acetate (100 mg/L) in the drinking water or distilled water for 14 days. After 14 days, lead acetate solution was substituted by water distilled for more 28 days, as control group. Systolic blood pressure (SBP) was measured weekly by tail plethysmography, and the vascular reactivity to phenylephrine in isolated aortic rings was evaluated at end of treatment time. The increase in SBP induced by lead was reversed after stopping exposure, and it was accompanied by a reduction on vasoconstrictor response to phenylephrine. L-NAME treatment increased the phenylephrine response in both groups, but its effect was greater in lead group. Our findings provide evidence that the increased modulation by NO on contractile response to phenylephrine could be a compensatory mechanism that might contribute to decrease blood pressure after lead treatment cessation.
Collapse
Affiliation(s)
- Gilson B Broseghini-Filho
- Department of Physiological Sciences, Federal University of Espirito Santo, Marechal Campos Ave, 1468, Maruípe, 29040-091, Vitória, ES, Brazil.
| | - Camila C Pereira Almenara
- Department of Physiological Sciences, Federal University of Espirito Santo, Marechal Campos Ave, 1468, Maruípe, 29040-091, Vitória, ES, Brazil
| | - Dalton V Vassallo
- Department of Physiological Sciences, Federal University of Espirito Santo, Marechal Campos Ave, 1468, Maruípe, 29040-091, Vitória, ES, Brazil
- Health Science Center of Vitoria, EMESCAM, Vitória, ES, Brazil
| | - Alessandra S Padilha
- Department of Physiological Sciences, Federal University of Espirito Santo, Marechal Campos Ave, 1468, Maruípe, 29040-091, Vitória, ES, Brazil
| |
Collapse
|
196
|
Adamopoulos C, Piperi C, Gargalionis AN, Dalagiorgou G, Spilioti E, Korkolopoulou P, Diamanti-Kandarakis E, Papavassiliou AG. Advanced glycation end products upregulate lysyl oxidase and endothelin-1 in human aortic endothelial cells via parallel activation of ERK1/2-NF-κB and JNK-AP-1 signaling pathways. Cell Mol Life Sci 2016; 73:1685-1698. [PMID: 26646068 PMCID: PMC11108501 DOI: 10.1007/s00018-015-2091-z] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/26/2015] [Accepted: 11/09/2015] [Indexed: 10/22/2022]
Abstract
Endothelial dysfunction involves deregulation of the key extracellular matrix (ECM) enzyme lysyl oxidase (LOX) and the vasoconstrictor protein, endothelin-1 (ET-1), whose gene expression can be modulated by the transcriptional activators nuclear factor kappa B (NF-κB) and activator protein-1 (AP-1). Advanced glycation end products (AGEs) present an aggravating factor of endothelial dysfunction which upon engagement to their receptor RAGE induce upregulation of mitogen-activated protein kinases (MAPKs), leading to NF-κB and AP-1 potentiation. We hypothesized that AGEs could induce NF-κΒ- and AP-1-dependent regulation of LOX and ET-1 expression via the AGE/RAGE/MAPK signaling axis. Western blot, real-time qRT-PCR, FACS analysis and electrophoretic mobility-shift assays were employed in human aortic endothelial cells (HAECs) following treatment with AGE-bovine serum albumin (AGE-BSA) to investigate the signaling pathway towards this hypothesis. Furthermore, immunohistochemical analysis of AGEs, RAGE, LOX and ET-1 expression was conducted in aortic endothelium of a rat experimental model exposed to high- or low-AGE content diet. HAECs exposed to AGE-BSA for various time points exhibited upregulation of LOX and ET-1 mRNA levels in a dose- and time-dependent manner. Exposure of HAECs to AGE-BSA also showed specific elevation of phospho(p)-ERK1/2 and p-JNK levels in a dose- and time-dependent fashion. AGE administration significantly increased NF-κΒ- and AP-1-binding activity to both LOX and ET-1 cognate promoter regions. Moreover, LOX and ET-1 overexpression in rat aortic endothelium upon high-AGE content diet confirmed the functional interrelation of these molecules. Our findings demonstrate that AGEs trigger NF-κΒ- and AP-1-mediated upregulation of LOX and ET-1 via the AGE/RAGE/MAPK signaling cascade in human endothelial cells, thus contributing to distorted endothelial homeostasis by impairing endothelial barrier function, altering ECM biomechanical properties and cell proliferation.
Collapse
Affiliation(s)
- Christos Adamopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75, M. Asias Street, 11527, Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75, M. Asias Street, 11527, Athens, Greece
| | - Antonios N Gargalionis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75, M. Asias Street, 11527, Athens, Greece
| | - Georgia Dalagiorgou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75, M. Asias Street, 11527, Athens, Greece
| | - Eliana Spilioti
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75, M. Asias Street, 11527, Athens, Greece
| | - Penelope Korkolopoulou
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527, Athens, Greece
| | - Evanthia Diamanti-Kandarakis
- Third Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, 'Sotiria' Hospital, 11527, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75, M. Asias Street, 11527, Athens, Greece.
| |
Collapse
|
197
|
Vilela-Martin JF, Giollo-Junior LT, Chiappa GR, Cipriano-Junior G, Vieira PJC, dos Santos Ricardi F, Paz-Landim MI, de Andrade DO, Cestário EDES, Cosenso-Martin LN, Yugar-Toledo JC, Cipullo JP. Effects of transcutaneous electrical nerve stimulation (TENS) on arterial stiffness and blood pressure in resistant hypertensive individuals: study protocol for a randomized controlled trial. Trials 2016; 17:168. [PMID: 27026087 PMCID: PMC4812656 DOI: 10.1186/s13063-016-1302-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 03/18/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Resistant hypertension (RH) treatment requires an adequate and intense therapeutic approach. However, the results are not always satisfactory despite intensive treatment. Of the different pathophysiological mechanisms involved in the pathogenesis of RH, sympathetic overstimulation and therapies that block the sympathetic system have been widely studied. These approaches, however, are invasive and expensive. Another possible approach is by transcutaneous electrical nerve stimulation (TENS), a noninvasive method that modulates activity by using low-frequency transcutaneous electrical stimulation to inhibit primary afferent pathways. Thus, the current study will evaluate the effect of applying TENS in the cervicothoracic region of subjects with RH and will seek to develop a new low-cost and readily available therapy to treat this group of hypertensive individuals. METHODS/DESIGN This is a randomized, single blind (subject), parallel-assignment study controlled with a sham group and including participants aged 40 to 70 years with resistant hypertension. The trial has two arms: the treatment and control (sham group). The treatment group will be submitted to the stimulation procedure (TENS). The sham group will not be submitted to stimulation. The primary outcomes will be a reduction in the peripheral blood pressure and adverse events. The secondary outcomes will be a reduction the central blood pressure. The study will last 30 days. The sample size was calculated assuming an alpha error of 5 % to reject the null hypothesis with a statistical power of 80 %, thereby resulting in 28 participants per group (intervention versus sham). DISCUSSION In recent decades, RH has become very common and costly. Adequate control requires several drugs, and in many cases, treatment is not successful. Sympathetic nervous system inhibition by renal denervation and central inhibition have significant effects in reducing BP; however, these treatments are costly and invasive. Another type of sympathetic nervous system inhibition can also be noninvasively achieved by electric current. Therefore, the application of TENS may be a new therapeutic option for treating resistant hypertensive individuals. TRIAL REGISTRATION Clinical Trials NCT02365974.
Collapse
Affiliation(s)
- José Fernando Vilela-Martin
- />Internal Medicine Department and Hospital de Base, Hypertension Clinic, Medical School in São José do Rio Preto (FAMERP), Av Anísio Haddad 7700 casa 129, Jd das Palmeiras, 15093-000 São José do Rio Preto, SP Brazil
| | - Luiz Tadeu Giollo-Junior
- />Internal Medicine Department and Hospital de Base, Hypertension Clinic, Medical School in São José do Rio Preto (FAMERP), Av Anísio Haddad 7700 casa 129, Jd das Palmeiras, 15093-000 São José do Rio Preto, SP Brazil
| | - Gaspar Rogério Chiappa
- />Cardiology Division, Federal University of Rio Grande do Sul (UFRS), Porto Alegre, Brazil
| | | | | | - Fábio dos Santos Ricardi
- />Internal Medicine Department and Hospital de Base, Hypertension Clinic, Medical School in São José do Rio Preto (FAMERP), Av Anísio Haddad 7700 casa 129, Jd das Palmeiras, 15093-000 São José do Rio Preto, SP Brazil
| | - Manoel Ildefonso Paz-Landim
- />Internal Medicine Department and Hospital de Base, Hypertension Clinic, Medical School in São José do Rio Preto (FAMERP), Av Anísio Haddad 7700 casa 129, Jd das Palmeiras, 15093-000 São José do Rio Preto, SP Brazil
| | - Days Oliveira de Andrade
- />Internal Medicine Department and Hospital de Base, Hypertension Clinic, Medical School in São José do Rio Preto (FAMERP), Av Anísio Haddad 7700 casa 129, Jd das Palmeiras, 15093-000 São José do Rio Preto, SP Brazil
| | - Elizabeth do Espírito Santo Cestário
- />Internal Medicine Department and Hospital de Base, Hypertension Clinic, Medical School in São José do Rio Preto (FAMERP), Av Anísio Haddad 7700 casa 129, Jd das Palmeiras, 15093-000 São José do Rio Preto, SP Brazil
| | - Luciana Neves Cosenso-Martin
- />Internal Medicine Department and Hospital de Base, Hypertension Clinic, Medical School in São José do Rio Preto (FAMERP), Av Anísio Haddad 7700 casa 129, Jd das Palmeiras, 15093-000 São José do Rio Preto, SP Brazil
| | - Juan Carlos Yugar-Toledo
- />Internal Medicine Department and Hospital de Base, Hypertension Clinic, Medical School in São José do Rio Preto (FAMERP), Av Anísio Haddad 7700 casa 129, Jd das Palmeiras, 15093-000 São José do Rio Preto, SP Brazil
| | - José Paulo Cipullo
- />Internal Medicine Department and Hospital de Base, Hypertension Clinic, Medical School in São José do Rio Preto (FAMERP), Av Anísio Haddad 7700 casa 129, Jd das Palmeiras, 15093-000 São José do Rio Preto, SP Brazil
| |
Collapse
|
198
|
Romero M, Leon-Gomez E, Lobysheva I, Rath G, Dogné JM, Feron O, Dessy C. Effects of BM-573 on Endothelial Dependent Relaxation and Increased Blood Pressure at Early Stages of Atherosclerosis. PLoS One 2016; 11:e0152579. [PMID: 27019366 PMCID: PMC4809599 DOI: 10.1371/journal.pone.0152579] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 03/16/2016] [Indexed: 12/19/2022] Open
Abstract
Endothelial dysfunction is considered to be an early event in atherosclerosis and plays a pivotal role in the development, progression and clinical complications of atherosclerosis. Previous studies have shown the beneficial effects of combined inhibition of thromboxane synthase and antagonism of thromboxane receptors by BM-573 on atherosclerosis; however our knowledge about the beneficial effects of BM-573 on endothelial function and increased blood pressure related to early stage of atherosclerosis is limited. In the present study, we investigated the effects of short-term (3 μM, 1 hour) and chronic (10 mg/L, 8 weeks) treatments with BM-573 on vasodilatory function, nitric oxide (NO) bioavailability, oxidative stress and systolic blood pressure in 15 weeks old apolipoprotein E-deficient (ApoE-KO) mice. ApoE-KO mice showed a reduced endothelium-derived relaxation. In addition, NO bioavailability was reduced and oxidative stress and blood pressure were increased in ApoE-KO mice versus wild-type mice. BM-573 treatments were able to improve the relaxation profile in ApoE-KO mice. Short-term effects of BM-573 were mainly mediated by an increased phosphorylation of both eNOS and Akt, whereas BM-573 in vivo treatment also reduced oxidative stress and restored NO bioavailability. In addition, chronic administration of BM-573 reduced systolic blood pressure in ApoE-KO mice. In conclusion, pharmacological modulation of TxA2 biosynthesis and biological activities by dual TP antagonism/TxAS inhibition with BM-573, already known to prevent plaque formation, has the potential to correct vasodilatory dysfunction at the early stages of atherosclerosis.
Collapse
Affiliation(s)
- Miguel Romero
- Pole of Pharmacology and Therapeutics (FATH), Institute of Experimental & Clinical Research (IREC), Université Catholique de Louvain (UCL) Medical School, Brussels, Belgium
- * E-mail: (MR); (CD)
| | - Elvira Leon-Gomez
- Pole of Pharmacology and Therapeutics (FATH), Institute of Experimental & Clinical Research (IREC), Université Catholique de Louvain (UCL) Medical School, Brussels, Belgium
| | - Irina Lobysheva
- Pole of Pharmacology and Therapeutics (FATH), Institute of Experimental & Clinical Research (IREC), Université Catholique de Louvain (UCL) Medical School, Brussels, Belgium
| | - Géraldine Rath
- Pole of Pharmacology and Therapeutics (FATH), Institute of Experimental & Clinical Research (IREC), Université Catholique de Louvain (UCL) Medical School, Brussels, Belgium
| | | | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institute of Experimental & Clinical Research (IREC), Université Catholique de Louvain (UCL) Medical School, Brussels, Belgium
| | - Chantal Dessy
- Pole of Pharmacology and Therapeutics (FATH), Institute of Experimental & Clinical Research (IREC), Université Catholique de Louvain (UCL) Medical School, Brussels, Belgium
- * E-mail: (MR); (CD)
| |
Collapse
|
199
|
Deer RR, Stallone JN. Effects of estrogen on cerebrovascular function: age-dependent shifts from beneficial to detrimental in small cerebral arteries of the rat. Am J Physiol Heart Circ Physiol 2016; 310:H1285-94. [PMID: 26993224 DOI: 10.1152/ajpheart.00645.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 03/14/2016] [Indexed: 02/06/2023]
Abstract
In the present study, interactions of age and estrogen in the modulation of cerebrovascular function were examined in small arteries <150 μM. The hypothesis tested was that age enhances deleterious effects of exogenous estrogen by augmenting constrictor prostanoid (CP)-potentiated reactivity of the female (F) cerebrovasculature. F Sprague-Dawley rats approximating key stages of "hormonal aging" in humans were studied: perimenopausal (mature multi-gravid, MA, cyclic, 5-6 mo of age) and postmenopausal (reproductively senescent, RS, acyclic 10-12 mo of age). Rats underwent bilateral ovariectomy and were given estrogen replacement therapy (E) or placebo (O) for 14-21 days. Vasopressin reactivity (VP, 10(-12)-10(-7) M) was measured in pressurized middle cerebral artery segments, alone or in the presence of COX-1- (SC560, 1 μM) or COX-2- (NS398, 10 μM) selective inhibitors. VP-stimulated release of prostacyclin (PGI2) and thromboxane (TXA2) were assessed by radioimmunoassay of 6-keto-PGF1α and TXB2 (stable metabolites). VP-induced vasoconstriction was attenuated in ovariectomized + estrogen-replaced, multigravid adult rats (5-6 mo; MAE) but potentiated in older ovariectomized + estrogen-replaced, reproductively senescent rats (12-14 mo; RSE). SC560 and NS398 reduced reactivity similarly in ovariectomized multigravid adult rats (5-6 mo; MAO) and ovariectomized reproductively senescent rat (12-14 mo; RSO). In MAE, reactivity to VP was reduced to a greater extent by SC560 than by NS398; however, in RSE, this effect was reversed. VP-stimulated PGI2 was increased by estrogen, yet reduced by age. VP-stimulated TXA2 was increased by estrogen and age in RSE but did not differ in MAO and RSO. Taken together, these data reveal that the vascular effects of estrogen are distinctly age-dependent in F rats. In younger MA, beneficial and protective effects of estrogen are evident (decreased vasoconstriction, increased dilator prostanoid function). Conversely, in older RS, detrimental effects of estrogen begin to be manifested (enhanced vasoconstriction and CP function). These findings may lead to age-specific estrogen replacement therapies that maximize beneficial and minimize detrimental effects of this hormone on small cerebral arteries that regulate blood flow.
Collapse
Affiliation(s)
- Rachel R Deer
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas; and
| | - John N Stallone
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas; and Women's Health Division, Michael E. DeBakey Institute, Texas A&M University, College Station, Texas
| |
Collapse
|
200
|
Romaschenko VP, Zinovkin RA, Galkin II, Zakharova VV, Panteleeva AA, Tokarchuk AV, Lyamzaev KG, Pletjushkina OY, Chernyak BV, Popova EN. Low Concentrations of Uncouplers of Oxidative Phosphorylation Prevent Inflammatory Activation of Endothelial Cells by Tumor Necrosis Factor. BIOCHEMISTRY (MOSCOW) 2016; 80:610-9. [PMID: 26071781 DOI: 10.1134/s0006297915050144] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In endothelial cells, mitochondria play an important regulatory role in physiology as well as in pathophysiology related to excessive inflammation. We have studied the effect of low doses of mitochondrial uncouplers on inflammatory activation of endothelial cells using the classic uncouplers 2,4-dinitrophenol and 4,5,6,7-tetrachloro-2-trifluoromethylbenzimidazole, as well as the mitochondria-targeted cationic uncoupler dodecyltriphenylphosphonium (C12TPP). All of these uncouplers suppressed the expression of E-selectin, adhesion molecules ICAM1 and VCAM1, as well as the adhesion of neutrophils to endothelium induced by tumor necrosis factor (TNF). The antiinflammatory action of the uncouplers was at least partially mediated by the inhibition of NFκB activation due to a decrease in phosphorylation of the inhibitory subunit IκBα. The dynamic concentration range for the inhibition of ICAM1 expression by C12TPP was three orders of magnitude higher compared to the classic uncouplers. Probably, the decrease in membrane potential inhibited the accumulation of penetrating cations into mitochondria, thus lowering the uncoupling activity and preventing further loss of mitochondrial potential. Membrane potential recovery after the removal of the uncouplers did not abolish its antiinflammatory action. Thus, mild uncoupling could induce TNF resistance in endothelial cells. We found no significant stimulation of mitochondrial biogenesis or autophagy by the uncouplers. However, we observed a decrease in the relative amount of fragmented mitochondria. The latter may significantly change the signaling properties of mitochondria. Earlier we showed that both classic and mitochondria-targeted antioxidants inhibited the TNF-induced NFκB-dependent activation of endothelium. The present data suggest that the antiinflammatory effect of mild uncoupling is related to its antioxidant action.
Collapse
Affiliation(s)
- V P Romaschenko
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, 119991, Russia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|