1
|
Zambolin F, Giuriato G, Laginestra FG, Ottaviani MM, Favaretto T, Calabria E, Duro-Ocana P, Bagley L, Faisal A, Peçanha T, McPhee JS, Venturelli M. Effects of nociceptive and mechanosensitive afferents sensitization on central and peripheral hemodynamics following exercise-induced muscle damage. J Appl Physiol (1985) 2022; 133:945-958. [PMID: 35981730 DOI: 10.1152/japplphysiol.00302.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/04/2022] [Accepted: 08/16/2022] [Indexed: 11/22/2022] Open
Abstract
This study aims to test the separated and combined effects of mechanoreflex activation and nociception through exercise-induced muscle damage (EIMD) on central and peripheral hemodynamics before and during single passive leg movement (sPLM). Eight healthy young males undertook four experimental sessions, in which a sPLM was performed on the dominant limb while in each specific session the contralateral was: 1) in a resting condition (CTRL), 2) stretched (ST), 3) resting after EIMD called delayed onset muscle soreness (DOMS) condition, or 4) stretched after EIMD (DOMS + ST). EIMD was used to induce DOMS in the following 24-48 h. Femoral blood flow (FBF) was assessed using Doppler ultrasound whereas central hemodynamics were assessed via finger photoplethysmography. Leg vascular conductance (LVC) was calculated as FBF/mean arterial pressure (MAP). RR-intervals were analyzed in the time (root mean squared of successive intervals; RMSSD) and frequency domain [low frequency (LF)/high frequency (HF)]. Blood samples were collected before each condition and gene expression analysis showed increased fold changes for P2X4 and IL1β in DOMS and DOMS + ST compared with baseline. Resting FBF and LVC were decreased only in the DOMS + ST condition (-26 mL/min and -50 mL/mmHg/min respectively) with decreased RMSSD and increased LF/HF ratio. MAP, HR, CO, and SV were increased in ST and DOMS + ST compared with CTRL. Marked decreases of Δpeaks and AUC were observed for FBF (Δ: -146 mL/min and -265 mL respectively) and LVC (Δ: -8.66 mL/mmHg/min and ±1.7 mL/mmHg/min respectively) all P < 0.05. These results suggest that the combination of mechanoreflex and nociception resulted in decreased vagal tone and concomitant rise in sympathetic drive that led to increases in resting central hemodynamics with reduced limb blood flow before and during sPLM.NEW & NOTEWORTHY Exercise-induced muscle damage (EIMD) is a well-known model to study mechanical hyperalgesia and muscle peripheral nerve sensitizations. The combination of static stretching protocol on the damaged limb extensively increases resting central hemodynamics with reduction in resting limb blood flow and passive leg movement-induced hyperemia. The mechanism underlining these results may be linked to reduction of vagal tone with concomitant increase in sympathetic activity following mechano- and nociceptive activation.
Collapse
Affiliation(s)
- Fabio Zambolin
- Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester, United Kingdom
- Manchester Metropolitan University Institute of Sport, Manchester Metropolitan University, Manchester, United Kingdom
| | - Gaia Giuriato
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Fabio Giuseppe Laginestra
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Matteo Maria Ottaviani
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- Department Medicine, University of Udine, Udine, Italy
| | - Thomas Favaretto
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- Department of Neurosurgery, University Politecnica delle Marche, Ancona, Italy
| | - Elisa Calabria
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Pablo Duro-Ocana
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
- Department of Anesthesia, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Liam Bagley
- Manchester Metropolitan University Institute of Sport, Manchester Metropolitan University, Manchester, United Kingdom
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
- Department of Anesthesia, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Azmy Faisal
- Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester, United Kingdom
- Manchester Metropolitan University Institute of Sport, Manchester Metropolitan University, Manchester, United Kingdom
- Faculty of Physical Education for Men, Alexandria University, Alexandria, Egypt
| | - Tiago Peçanha
- Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester, United Kingdom
- Manchester Metropolitan University Institute of Sport, Manchester Metropolitan University, Manchester, United Kingdom
| | - Jamie Stewart McPhee
- Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester, United Kingdom
- Manchester Metropolitan University Institute of Sport, Manchester Metropolitan University, Manchester, United Kingdom
| | - Massimo Venturelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| |
Collapse
|
2
|
Hansen FB, Esteves GV, Mogensen S, Prat-Duran J, Secher N, Løfgren B, Granfeldt A, Simonsen U. Increased cerebral endothelium-dependent vasodilation in rats in the postcardiac arrest period. J Appl Physiol (1985) 2021; 131:1311-1327. [PMID: 34435510 DOI: 10.1152/japplphysiol.00373.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cardiovascular lability is common after cardiac arrest. We investigated whether altered endothelial function is present in cerebral and mesenteric arteries 2 and 4 h after resuscitation. Male Sprague-Dawley rats were anesthetized, intubated, ventilated, and intravascularly catheterized whereupon rats were randomized into four groups. Following 7 min of asphyxial cardiac arrest and subsequent resuscitation, cardiac arrest and sham rats were observed for either 2 or 4 h. Neuron-specific enolase levels were measured in blood samples. Middle cerebral artery segments and small mesenteric arteries were isolated and examined in microvascular myographs. qPCR and immunofluorescence analysis were performed on cerebral arteries. In cerebral arteries, bradykinin-induced vasodilation was inhibited in the presence of either calcium-activated K+ channel blockers (UCL1684 and senicapoc) or the nitric oxide (NO) synthase inhibitor, Nω-nitro-L-arginine methyl ester hydrochloride (l-NAME), whereas the combination abolished bradykinin-induced vasodilation across groups. Neuron-specific enolase levels were significantly increased in cardiac arrest rats. Cerebral vasodilation was comparable between the 2-h groups, but markedly enhanced in response to bradykinin, NS309 (an opener of small and intermediate calcium-activated K+ channels), and sodium nitroprusside 4 h after cardiac arrest. Endothelial NO synthase and guanylyl cyclase subunit α-1 mRNA expression was unaltered after 2 h, but significantly decreased 4 h after resuscitation. In mesenteric arteries, the endothelium-dependent vasodilation was comparable between corresponding groups at both 2 and 4 h. Our findings show enhanced cerebral endothelium-dependent vasodilation 4 h after cardiac arrest mediated by potentiated endothelial-derived hyperpolarization and NO pathways. Altered cerebral endothelium-dependent vasodilation may contribute to disturbed cerebral perfusion after cardiac arrest.NEW & NOTEWORTHY This is the first study, to our knowledge, to demonstrate enhanced endothelium-dependent vasodilation in middle cerebral arteries in a cardiac arrest rat model. The increased endothelium-dependent vasodilation was a result of potentiated endothelium-derived hyperpolarization and endothelial nitric oxide pathways. Immunofluorescence microscopy confirmed the presence of relevant receptors and eNOS in cerebral arteries, whereas qPCR showed altered expression of genes related to guanylyl cyclase and eNOS. Altered endothelium-dependent vasoregulation may contribute to disturbed cerebral blood flow in the postcardiac arrest period.
Collapse
Affiliation(s)
- Frederik Boe Hansen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Susie Mogensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Niels Secher
- Department of Anesthesiology and Intensive Care, Aarhus University Hospital, Aarhus, Denmark
| | - Bo Løfgren
- Research Center for Emergency Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Asger Granfeldt
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Anesthesiology and Intensive Care, Aarhus University Hospital, Aarhus, Denmark
| | - Ulf Simonsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
3
|
Endothelium-Dependent Hyperpolarization (EDH) in Diabetes: Mechanistic Insights and Therapeutic Implications. Int J Mol Sci 2019; 20:ijms20153737. [PMID: 31370156 PMCID: PMC6695796 DOI: 10.3390/ijms20153737] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/25/2019] [Accepted: 07/30/2019] [Indexed: 02/07/2023] Open
Abstract
Diabetes mellitus is one of the major risk factors for cardiovascular disease and is an important health issue worldwide. Long-term diabetes causes endothelial dysfunction, which in turn leads to diabetic vascular complications. Endothelium-derived nitric oxide is a major vasodilator in large-size vessels, and the hyperpolarization of vascular smooth muscle cells mediated by the endothelium plays a central role in agonist-mediated and flow-mediated vasodilation in resistance-size vessels. Although the mechanisms underlying diabetic vascular complications are multifactorial and complex, impairment of endothelium-dependent hyperpolarization (EDH) of vascular smooth muscle cells would contribute at least partly to the initiation and progression of microvascular complications of diabetes. In this review, we present the current knowledge about the pathophysiology and underlying mechanisms of impaired EDH in diabetes in animals and humans. We also discuss potential therapeutic approaches aimed at the prevention and restoration of EDH in diabetes.
Collapse
|
4
|
Sena CM, Leandro A, Azul L, Seiça R, Perry G. Vascular Oxidative Stress: Impact and Therapeutic Approaches. Front Physiol 2018; 9:1668. [PMID: 30564132 PMCID: PMC6288353 DOI: 10.3389/fphys.2018.01668] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 11/06/2018] [Indexed: 12/13/2022] Open
Abstract
Oxidative stress has been defined as an imbalance between oxidants and antioxidants and more recently as a disruption of redox signaling and control. It is generally accepted that oxidative stress can lead to cell and tissue injury having a fundamental role in vascular dysfunction. Physiologically, reactive oxygen species (ROS) control vascular function by modulating various redox-sensitive signaling pathways. In vascular disorders, oxidative stress instigates endothelial dysfunction and inflammation, affecting several cells in the vascular wall. Vascular ROS are derived from multiple sources herein discussed, which are prime targets for therapeutic development. This review focuses on oxidative stress in vascular physiopathology and highlights different strategies to inhibit ROS production.
Collapse
Affiliation(s)
- Cristina M. Sena
- Institute of Physiology, Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Adriana Leandro
- Institute of Physiology, Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Lara Azul
- Institute of Physiology, Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Raquel Seiça
- Institute of Physiology, Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - George Perry
- College of Sciences, One UTSA Circle, University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
5
|
SKA-31, an activator of endothelial Ca 2+-activated K + channels evokes robust vasodilation in rat mesenteric arteries. Eur J Pharmacol 2018; 831:60-67. [PMID: 29753043 DOI: 10.1016/j.ejphar.2018.05.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/04/2018] [Accepted: 05/08/2018] [Indexed: 12/17/2022]
Abstract
It is now well recognized that endothelial KCa2.3 and KCa3.1 channel activities contribute to dilation of resistance arteries via endothelium-mediated hyperpolarization and vascular smooth muscle relaxation. In this study, we have investigated the functional effect of the KCa channel activator SKA-31 in third order rat mesenteric arteries using arterial pressure myography. Isolated arteries were cannulated, pressurized intraluminally to 70 mmHg at 36 °C and then constricted with 1 μM phenylephrine. Acute bath exposure to SKA-31 evoked a robust and reversible inhibition of developed tone (IC50 = 0.22 μM). The vasodilatory effects of SKA-31 and acetylcholine were blunted in the presence of KCa2.3 and KCa3.1 channel antagonists, and were largely prevented following endothelial denudation. Western blot and q-PCR analyses of isolated mesenteric arteries revealed KCa2.3 and KCa3.1 channel expression at the protein and mRNA levels, respectively. Penitrem-A, an inhibitor of KCa1.1 channels, decreased vasodilatory responses to acetylcholine, sodium nitroprusside and NS-1619, but had little effect on SKA-31. Similarly, bath exposure to the eNOS inhibitor L-NAME did not alter SKA-31 and acetylcholine-mediated vasodilation. Collectively, these data highlight the major cellular mechanisms by which the endothelial KCa channel activator SKA-31 inhibits agonist-evoked vasoconstriction in rat small mesenteric arteries.
Collapse
|
6
|
Fernández-Alfonso MS, Somoza B, Tsvetkov D, Kuczmanski A, Dashwood M, Gil-Ortega M. Role of Perivascular Adipose Tissue in Health and Disease. Compr Physiol 2017; 8:23-59. [PMID: 29357124 DOI: 10.1002/cphy.c170004] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Perivascular adipose tissue (PVAT) is cushion of fat tissue surrounding blood vessels, which is phenotypically different from other adipose tissue depots. PVAT is composed of adipocytes and stromal vascular fraction, constituted by different populations of immune cells, endothelial cells, and adipose-derived stromal cells. It expresses and releases an important number of vasoactive factors with paracrine effects on vascular structure and function. In healthy individuals, these factors elicit a net anticontractile and anti-inflammatory paracrine effect aimed at meeting hemodynamic and metabolic demands of specific organs and regions of the body. Pathophysiological situations, such as obesity, diabetes or hypertension, induce changes in its amount and in the expression pattern of vasoactive factors leading to a PVAT dysfunction in which the beneficial paracrine influence of PVAT is shifted to a pro-oxidant, proinflammatory, contractile, and trophic environment leading to functional and structural cardiovascular alterations and cardiovascular disease. Many different PVATs surrounding a variety of blood vessels have been described and exhibit regional differences. Both protective and deleterious influence of PVAT differs regionally depending on the specific vascular bed contributing to variations in the susceptibility of arteries and veins to vascular disease. PVAT therefore, might represent a novel target for pharmacological intervention in cardiovascular disease. © 2018 American Physiological Society. Compr Physiol 8:23-59, 2018.
Collapse
Affiliation(s)
| | - Beatriz Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - Dmitry Tsvetkov
- Department of Anestesiology, Perioperative and Pain Medicine, HELIOS Klinikum, Berlin-Buch GmbH, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, Department of Pharmacology and Experimental Therapy, Eberhard Karls University Hospitals and Clinics, and Interfaculty Center of Pharmacogenomics and Drug Research, Tübingen, Germany
| | - Artur Kuczmanski
- Department of Anestesiology, Perioperative and Pain Medicine, HELIOS Klinikum, Berlin-Buch GmbH, Germany
| | - Mick Dashwood
- Royal Free Hospital Campus, University College Medical School, London, United Kingdom
| | - Marta Gil-Ortega
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| |
Collapse
|
7
|
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: 103] [Impact Index Per Article: 12.9] [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
|
8
|
Diniz MC, Olivon VC, Tavares LD, Simplicio JA, Gonzaga NA, de Souza DG, Bendhack LM, Tirapelli CR, Bonaventura D. Mechanisms underlying sodium nitroprusside-induced tolerance in the mouse aorta: Role of ROS and cyclooxygenase-derived prostanoids. Life Sci 2017; 176:26-34. [DOI: 10.1016/j.lfs.2017.03.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 03/15/2017] [Accepted: 03/20/2017] [Indexed: 01/15/2023]
|
9
|
Verhoeven F, Totoson P, Maguin-Gaté K, Prigent-Tessier A, Marie C, Wendling D, Moretto J, Prati C, Demougeot C. Glucocorticoids improve endothelial function in rheumatoid arthritis: a study in rats with adjuvant-induced arthritis. Clin Exp Immunol 2017; 188:208-218. [PMID: 28152574 DOI: 10.1111/cei.12938] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2017] [Indexed: 12/15/2022] Open
Abstract
To determine the effect of glucocorticoids (GCs) on endothelial dysfunction (ED) and on traditional cardiovascular (CV) risk factors in the adjuvant-induced arthritis (AIA) rat model. At the first signs of AIA, a high dose (HD) [10 mg/kg/day, intraperitoneally (i.p.), GC-HD] or low dose (LD) (1 mg/kg/day, i.p., GC-LD) of prednisolone was administered for 3 weeks. Endothelial function was studied in aortic rings relaxed with acetylcholine (Ach) with or without inhibitors of nitric oxide synthase (NOS), cyclooxygenase 2 (COX-2), arginase, endothelium derived hyperpolarizing factor (EDHF) and superoxide anions ( O2-°) production. Aortic expression of endothelial NOS (eNOS), Ser1177-phospho-eNOS, COX-2, arginase-2, p22phox and p47phox was evaluated by Western blotting analysis. Arthritis scores, blood pressure, heart rate and blood levels of cytokines, triglycerides, cholesterol and glucose were measured. GC-HD but not GC-LD reduced arthritis score significantly and improved Ach-induced relaxation (P < 0·05). The positive effect of GC-HD resulted from increased NOS activity and EDHF production and decreased COX-2/arginase activities and O2-° production. These functional effects relied upon increased phospho-eNOS expression and decreased COX-2, arginase-2 and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase expression. Despite the lack of effect of GC-LD on ED, it increased NOS and EDHF and down-regulated O2-° pathways but did not change arginase and COX-2 pathways. GC-HD increased triglycerides levels and blood pressure significantly (P < 0·05). Both doses of GCs decreased to the same extent as plasma interleukin (IL)-1β and tumour necrosis factor (TNF)-α levels (P < 0·05). Our data demonstrated that subchronic treatment with prednisolone improved endothelial function in AIA via pleiotropic effects on endothelial pathways. These effects occurred independently of the deleterious cardiometabolic effects and the impact of prednisolone on systemic inflammation.
Collapse
Affiliation(s)
- F Verhoeven
- PEPITE EA4267, FHU INCREASE, Université Bourgogne Franche-Comté, Besançon, France.,Service de Rhumatologie, CHRU Besançon, France
| | - P Totoson
- PEPITE EA4267, FHU INCREASE, Université Bourgogne Franche-Comté, Besançon, France
| | - K Maguin-Gaté
- PEPITE EA4267, FHU INCREASE, Université Bourgogne Franche-Comté, Besançon, France
| | | | - C Marie
- INSERM U1093, Université Bourgogne Franche-Comté, Dijon, France
| | - D Wendling
- Service de Rhumatologie, CHRU Besançon, France.,EA 4266, Université Bourgogne Franche-Comté, Besançon, France
| | - J Moretto
- PEPITE EA4267, FHU INCREASE, Université Bourgogne Franche-Comté, Besançon, France
| | - C Prati
- PEPITE EA4267, FHU INCREASE, Université Bourgogne Franche-Comté, Besançon, France.,Service de Rhumatologie, CHRU Besançon, France
| | - C Demougeot
- PEPITE EA4267, FHU INCREASE, Université Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|
10
|
Affiliation(s)
- Maik Gollasch
- Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, and Experimental and Clinical Research Center, a joint cooperation of the Charité – University Medicine Berlin and Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany;
| |
Collapse
|
11
|
Tanaka M. Study on the Physiological Benefits of Food Compounds on Vascular Health and Their Underlying Mechanisms. J JPN SOC FOOD SCI 2017. [DOI: 10.3136/nskkk.64.285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Mitsuru Tanaka
- Faculty of Agriculture, Graduate School of Kyushu University
| |
Collapse
|
12
|
Inflammatory Action of Secretory Phospholipases A2 from Snake Venoms. TOXINS AND DRUG DISCOVERY 2017. [DOI: 10.1007/978-94-007-6452-1_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
13
|
Bruder-Nascimento T, Ferreira NS, Zanotto CZ, Ramalho F, Pequeno IO, Olivon VC, Neves KB, Alves-Lopes R, Campos E, Silva CAA, Fazan R, Carlos D, Mestriner FL, Prado D, Pereira FV, Braga T, Luiz JPM, Cau SB, Elias PC, Moreira AC, Câmara NO, Zamboni DS, Alves-Filho JC, Tostes RC. NLRP3 Inflammasome Mediates Aldosterone-Induced Vascular Damage. Circulation 2016; 134:1866-1880. [PMID: 27803035 DOI: 10.1161/circulationaha.116.024369] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 10/04/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND Inflammation is a key feature of aldosterone-induced vascular damage and dysfunction, but molecular mechanisms by which aldosterone triggers inflammation remain unclear. The NLRP3 inflammasome is a pivotal immune sensor that recognizes endogenous danger signals triggering sterile inflammation. METHODS We analyzed vascular function and inflammatory profile of wild-type (WT), NLRP3 knockout (NLRP3-/-), caspase-1 knockout (Casp-1-/-), and interleukin-1 receptor knockout (IL-1R-/-) mice treated with vehicle or aldosterone (600 µg·kg-1·d-1 for 14 days through osmotic mini-pump) while receiving 1% saline to drink. RESULTS Here, we show that NLRP3 inflammasome plays a central role in aldosterone-induced vascular dysfunction. Long-term infusion of aldosterone in mice resulted in elevation of plasma interleukin-1β levels and vascular abnormalities. Mice lacking the IL-1R or the inflammasome components NLRP3 and caspase-1 were protected from aldosterone-induced vascular damage. In vitro, aldosterone stimulated NLRP3-dependent interleukin-1β secretion by bone marrow-derived macrophages by activating nuclear factor-κB signaling and reactive oxygen species generation. Moreover, chimeric mice reconstituted with NLRP3-deficient hematopoietic cells showed that NLRP3 in immune cells mediates aldosterone-induced vascular damage. In addition, aldosterone increased the expression of NLRP3, active caspase-1, and mature interleukin-1β in human peripheral blood mononuclear cells. Hypertensive patients with hyperaldosteronism or normal levels of aldosterone exhibited increased activity of NLRP3 inflammasome, suggesting that the effect of hyperaldosteronism on the inflammasome may be mediated through high blood pressure. CONCLUSIONS Together, these data demonstrate that NLRP3 inflammasome, through activation of IL-1R, is critically involved in the deleterious vascular effects of aldosterone, placing NLRP3 as a potential target for therapeutic interventions in conditions with high aldosterone levels.
Collapse
MESH Headings
- Acetylcholine/pharmacology
- Aldosterone/pharmacology
- Animals
- Bone Marrow Cells/cytology
- Bone Marrow Transplantation
- Caspase 1/deficiency
- Caspase 1/genetics
- Humans
- Intercellular Adhesion Molecule-1/genetics
- Intercellular Adhesion Molecule-1/metabolism
- Interleukin-1beta/blood
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/metabolism
- Macrophages/cytology
- Macrophages/drug effects
- Macrophages/metabolism
- Male
- Mesenteric Arteries/drug effects
- Mesenteric Arteries/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- NF-kappa B/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/deficiency
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Nigericin/pharmacology
- Reactive Oxygen Species/metabolism
- Receptors, Interleukin-1/deficiency
- Receptors, Interleukin-1/genetics
- Signal Transduction/drug effects
- Vascular Diseases/chemically induced
Collapse
Affiliation(s)
- Thiago Bruder-Nascimento
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.).
| | - Nathanne S Ferreira
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Camila Z Zanotto
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Fernanda Ramalho
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Isabela O Pequeno
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Vania C Olivon
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Karla B Neves
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Rheure Alves-Lopes
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Eduardo Campos
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Carlos Alberto A Silva
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Rubens Fazan
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Daniela Carlos
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Fabiola L Mestriner
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Douglas Prado
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Felipe V Pereira
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Tarcio Braga
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Joao Paulo M Luiz
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Stefany B Cau
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Paula C Elias
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Ayrton C Moreira
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Niels O Câmara
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Dario S Zamboni
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Jose Carlos Alves-Filho
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.)
| | - Rita C Tostes
- From Department of Pharmacology (T.B.-N., N.S.F., C.Z.Z., F.R., I.O.P., V.C.O., K.B.N., R.A.-L., E.C., F.L.M., D.P., J.P.M.L., J.C.A.F., R.C.T.), Department of Physiology (C.A.A.S., R.F.), Department of Biochemistry and Immunology (D.C.), Department of Clinical Medicine, Division of Endocrinology (P.C.E., A.C.M.), and Department of Cell and Molecular Biology (D.S.Z.), Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil (F.V.P., T.B., N.O.); and Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil (S.B.C.).
| |
Collapse
|
14
|
Bussey CE, Withers SB, Aldous RG, Edwards G, Heagerty AM. Obesity-Related Perivascular Adipose Tissue Damage Is Reversed by Sustained Weight Loss in the Rat. Arterioscler Thromb Vasc Biol 2016; 36:1377-85. [DOI: 10.1161/atvbaha.116.307210] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 04/26/2016] [Indexed: 11/16/2022]
Affiliation(s)
- Charlotte E. Bussey
- From the Institute of Cardiovascular Sciences (C.E.B., S.B.W., R.G.A., A.M.H.), and Faculty of Life Sciences (G.E.), University of Manchester, Manchester, United Kingdom
| | - Sarah B. Withers
- From the Institute of Cardiovascular Sciences (C.E.B., S.B.W., R.G.A., A.M.H.), and Faculty of Life Sciences (G.E.), University of Manchester, Manchester, United Kingdom
| | - Robert G. Aldous
- From the Institute of Cardiovascular Sciences (C.E.B., S.B.W., R.G.A., A.M.H.), and Faculty of Life Sciences (G.E.), University of Manchester, Manchester, United Kingdom
| | - Gillian Edwards
- From the Institute of Cardiovascular Sciences (C.E.B., S.B.W., R.G.A., A.M.H.), and Faculty of Life Sciences (G.E.), University of Manchester, Manchester, United Kingdom
| | - Anthony M. Heagerty
- From the Institute of Cardiovascular Sciences (C.E.B., S.B.W., R.G.A., A.M.H.), and Faculty of Life Sciences (G.E.), University of Manchester, Manchester, United Kingdom
| |
Collapse
|
15
|
Withaferin A protects against palmitic acid-induced endothelial insulin resistance and dysfunction through suppression of oxidative stress and inflammation. Sci Rep 2016; 6:27236. [PMID: 27250532 PMCID: PMC4890118 DOI: 10.1038/srep27236] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 05/13/2016] [Indexed: 01/01/2023] Open
Abstract
Activation of inflammatory pathways via reactive oxygen species (ROS) by free fatty acids (FFA) in obesity gives rise to insulin resistance and endothelial dysfunction. Withaferin A (WA), possesses both antioxidant and anti-inflammatory properties and therefore would be a good strategy to suppress palmitic acid (PA)-induced oxidative stress and inflammation and hence, insulin resistance and dysfunction in the endothelium. Effect of WA on PA-induced insulin resistance in human umbilical vein endothelial cells (HUVECs) was determined by evaluating insulin signaling mechanisms whilst effect of this drug on PA-induced endothelial dysfunction was determined in acetylcholine-mediated relaxation in isolated rat aortic preparations. WA significantly inhibited ROS production and inflammation induced by PA. Furthermore, WA significantly decreased TNF-α and IL-6 production in endothelial cells by specifically suppressing IKKβ/NF-κβ phosphorylation. WA inhibited inflammation-stimulated IRS-1 serine phosphorylation and improved the impaired insulin PI3-K signaling, and restored the decreased nitric oxide (NO) production triggered by PA. WA also decreased endothelin-1 and plasminogen activator inhibitor type-1 levels, and restored the impaired endothelium-mediated vasodilation in isolated aortic preparations. These findings suggest that WA inhibited both ROS production and inflammation to restore impaired insulin resistance in cultured endothelial cells and improve endothelial dysfunction in rat aortic rings.
Collapse
|
16
|
Totoson P, Maguin-Gaté K, Nappey M, Wendling D, Demougeot C. Endothelial Dysfunction in Rheumatoid Arthritis: Mechanistic Insights and Correlation with Circulating Markers of Systemic Inflammation. PLoS One 2016; 11:e0146744. [PMID: 26761790 PMCID: PMC4711944 DOI: 10.1371/journal.pone.0146744] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/20/2015] [Indexed: 12/30/2022] Open
Abstract
Objectives To determine mechanisms involved in endothelial dysfunction (ED) during the course of arthritis and to investigate the link between cytokines, chemokines and osteoprotegerin. Approach and Results Experiments were conducted on aortic rings at day 4 (preclinical), day 11 (onset of disease), day 33 (acute disease) and day 90 (chronic disease) after adjuvant-induced arthritis (AIA) in Lewis rats. At day 4, the unique vascular abnormality was a reduced norepinephrine-induced constriction. At day 11, endothelial function assessed by the relaxation to acetylcholine was normal despite increased cyclo-oxygenase-2 activity (COX-2) and overproduction of superoxide anions that was compensated by increased nitric oxide synthase (NOS) activity. At day 33, ED apparition coincides with the normalization of NOS activity. At day 90, ED was only observed in rats with a persisting imbalance between endothelial NOS and COX-2 pathways and higher plasma levels of IL-1β and TNFα. Plasma levels of IL-1β, TNFα and MIP-1α negatively correlated with Ach-induced relaxation throughout the course of AIA. Conclusions Our data identified increased endothelial NOS activity as an important compensatory response that opposes the ED in the early arthritis. Thereafter, a cross-talk between endothelial COX-2/NOS pathways appears as an important element for the occurrence of ED. Our results encourage determining the clinical value of IL-1β, TNFα and MIP-1α as biomarkers of ED in RA.
Collapse
MESH Headings
- Acetylcholine/pharmacology
- Animals
- Arthritis, Experimental/blood
- Arthritis, Experimental/diagnostic imaging
- Arthritis, Experimental/physiopathology
- Arthritis, Rheumatoid/blood
- Arthritis, Rheumatoid/diagnostic imaging
- Arthritis, Rheumatoid/physiopathology
- Biomarkers/blood
- Chemokines/blood
- Cyclic N-Oxides/pharmacology
- Cyclooxygenase 2/metabolism
- Disease Progression
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/pathology
- Endothelium, Vascular/physiopathology
- Immunization
- Inflammation/blood
- Inflammation/complications
- Male
- NG-Nitroarginine Methyl Ester/pharmacology
- Nitric Oxide Synthase Type III/metabolism
- Nitrobenzenes/pharmacology
- Nitroprusside/pharmacology
- Osteoprotegerin/blood
- Radiography
- Rats, Inbred Lew
- Spin Labels
- Sulfonamides/pharmacology
- Superoxides/metabolism
- Time Factors
- Vasoconstriction/drug effects
- Vasodilation/drug effects
Collapse
Affiliation(s)
- Perle Totoson
- EA 4267 FDE, FHU INCREASE, Univ. Bourgogne Franche-Comté, Besançon, France
| | - Katy Maguin-Gaté
- EA 4267 FDE, FHU INCREASE, Univ. Bourgogne Franche-Comté, Besançon, France
| | - Maude Nappey
- EA 4267 FDE, FHU INCREASE, Univ. Bourgogne Franche-Comté, Besançon, France
| | - Daniel Wendling
- Service de Rhumatologie, CHRU Besançon, Besançon, France
- EA 4266, Univ. Bourgogne Franche-Comté, Besançon, France
| | - Céline Demougeot
- EA 4267 FDE, FHU INCREASE, Univ. Bourgogne Franche-Comté, Besançon, France
- * E-mail:
| |
Collapse
|
17
|
Liu P, Xie Q, Wei T, Chen Y, Chen H, Shen W. Activation of the NLRP3 inflammasome induces vascular dysfunction in obese OLETF rats. Biochem Biophys Res Commun 2015; 468:319-25. [DOI: 10.1016/j.bbrc.2015.10.105] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 10/20/2015] [Indexed: 10/22/2022]
|
18
|
Oriowo MA. Perivascular adipose tissue, vascular reactivity and hypertension. Med Princ Pract 2015; 24 Suppl 1:29-37. [PMID: 24503717 PMCID: PMC6489082 DOI: 10.1159/000356380] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Accepted: 10/09/2013] [Indexed: 12/13/2022] Open
Abstract
Most blood vessels are surrounded by a variable amount of adventitial adipose tissue, perivascular adipose tissue (PVAT), which was originally thought to provide mechanical support for the vessel. It is now known that PVAT secretes a number of bioactive substances including vascular endothelial growth factor, tumor necrosis factor-alpha (TNF-α), leptin, adiponectin, insulin-like growth factor, interleukin-6, plasminogen activator substance, resistin and angiotensinogen. Several studies have shown that PVAT significantly modulated vascular smooth muscle contractions induced by a variety of agonists and electrical stimulation by releasing adipocyte-derived relaxing (ADRF) and contracting factors. The identity of ADRF is not yet known. However, several vasodilators have been suggested including adiponectin, angiotensin 1-7, hydrogen sulfide and methyl palmitate. The anticontractile effect of PVAT is mediated through the activation of potassium channels since it is abrogated by inhibiting potassium channels. Hypertension is characterized by a reduction in the size and amount of PVAT and this is associated with the attenuated anticontractile effect of PVAT in hypertension. However, since a reduction in size and amount of PVAT and the attenuated anticontractile effect of PVAT were already evident in prehypertensive rats with no evidence of impaired release of ADRF, there is the possibility that the anticontractile effect of PVAT was not directly related to an altered function of the adipocytes per se. Hypertension is characterized by low-grade inflammation and infiltration of macrophages. One of the adipokines secreted by macrophages is TNF-α. It has been shown that exogenously administered TNF-α enhanced agonist-induced contraction of a variety of vascular smooth muscle preparations and reduced endothelium-dependent relaxation. Other procontractile factors released by the PVAT include angiotensin II and superoxide. It is therefore possible that the loss could be due to an increased amount of these proinflammatory and procontractile factors. More studies are definitely required to confirm this.
Collapse
Affiliation(s)
- Mabayoje A Oriowo
- Department of Pharmacology and Toxicology, Faculty of Medicine, Health Sciences Centre, Kuwait University, Jabriya, Kuwait
| |
Collapse
|
19
|
Vallejo S, Palacios E, Romacho T, Villalobos L, Peiró C, Sánchez-Ferrer CF. The interleukin-1 receptor antagonist anakinra improves endothelial dysfunction in streptozotocin-induced diabetic rats. Cardiovasc Diabetol 2014; 13:158. [PMID: 25518980 PMCID: PMC4276125 DOI: 10.1186/s12933-014-0158-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 11/21/2014] [Indexed: 01/06/2023] Open
Abstract
Background Endothelial dysfunction is a crucial early phenomenon in vascular diseases linked to diabetes mellitus and associated to enhanced oxidative stress. There is increasing evidence about the role for pro-inflammatory cytokines, like interleukin-1β (IL-1β), in developing diabetic vasculopathy. We aimed to determine the possible involvement of this cytokine in the development of diabetic endothelial dysfunction, analysing whether anakinra, an antagonist of IL-1 receptors, could reduce this endothelial alteration by interfering with pro-oxidant and pro-inflammatory pathways into the vascular wall. Results In control and two weeks evolution streptozotocin-induced diabetic rats, either untreated or receiving anakinra, vascular reactivity and NADPH oxidase activity were measured, respectively, in isolated rings and homogenates from mesenteric microvessels, while nuclear factor (NF)-κB activation was determined in aortas. Plasma levels of IL-1β and tumor necrosis factor (TNF)-α were measured by ELISA. In isolated mesenteric microvessels from control rats, two hours incubation with IL-1β (1 to 10 ng/mL) produced a concentration-dependent impairment of endothelium-dependent relaxations, which were mediated by enhanced NADPH oxidase activity via IL-1 receptors. In diabetic rats treated with anakinra (100 or 160 mg/Kg/day for 3 or 7 days before sacrifice) a partial improvement of diabetic endothelial dysfunction occurred, together with a reduction of vascular NADPH oxidase and NF-κB activation. Endothelial dysfunction in diabetic animals was also associated to higher activities of the pro-inflammatory enzymes cyclooxygenase (COX) and the inducible isoform of nitric oxide synthase (iNOS), which were markedly reduced after anakinra treatment. Circulating IL-1β and TNF-α levels did not change in diabetic rats, but they were lowered by anakinra treatment. Conclusions In this short-term model of type 1 diabetes, endothelial dysfunction is associated to an IL-1 receptor-mediated activation of vascular NADPH oxidase and NF-κB, as well as to vascular inflammation. Moreover, endothelial dysfunction, vascular oxidative stress and inflammation were reduced after anakinra treatment. Whether this mechanism can be extrapolated to a chronic situation or whether it may apply to diabetic patients remain to be established. However, it may provide new insights to further investigate the therapeutic use of IL-1 receptor antagonists to obtain vascular benefits in patients with diabetes mellitus and/or atherosclerosis.
Collapse
Affiliation(s)
- Susana Vallejo
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo 4, 29029, Madrid, Spain.
| | - Erika Palacios
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo 4, 29029, Madrid, Spain. .,Present address: Departamento de Ciencias de la Salud, Edificio CN208, Oficina O, Universidad de las Américas, Puebla, México.
| | - Tania Romacho
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo 4, 29029, Madrid, Spain. .,Present address: Paul Langerhans-Group, Integrative Physiology, German Diabetes Center, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany.
| | - Laura Villalobos
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo 4, 29029, Madrid, Spain.
| | - Concepción Peiró
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo 4, 29029, Madrid, Spain.
| | - Carlos F Sánchez-Ferrer
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo 4, 29029, Madrid, Spain.
| |
Collapse
|
20
|
Ismail B, Aboul-Fotouh S, Mansour AA, Shehata HH, Salman MI, Ibrahim EA, Hassan OA, Abdel-tawab AM. Behavioural, metabolic, and endothelial effects of the TNF-α suppressor thalidomide on rats subjected to chronic mild stress and fed an atherogenic diet. Can J Physiol Pharmacol 2014; 92:375-85. [DOI: 10.1139/cjpp-2013-0446] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
There is accumulating evidence suggesting that depression is a risk factor for cardiovascular diseases. This study aimed to examine the hypothesis that the proinflammatory cytokine TNF-α would partially explain the link between depression and atherosclerotic endothelial changes. Rats were distributed among 6 groups: (i) control group; (ii) group subjected to chronic mild stress (CMS); (iii) group fed a cholesterol–cholic acid–thiouracil (CCT diet); and (iv) CMS group fed the CCT diet and treated with the vehicle for 8 weeks. The last 2 groups were subjected to CMS–CCT and received thalidomide (THAL) or imipramine (IMIP). Rats were assessed behaviorally (sucrose preference, open field, and forced-swimming tests). TNF-α protein was assessed from the serum, aorta, and liver. Aortic TNF-α gene expression (assessed using RT–PCR), serum lipid profile, and insulin levels were measured. Endothelial function was assessed in isolated aortic rings. The THAL and IMIP groups showed ameliorated CMS–CCT-related behavioral changes. CMS–CCT-induced metabolic and endothelial dysfunctions were improved in the THAL group but were worsened in the IMIP group. RT–PCR showed a significant reduction of aortic TNF-α mRNA expression in the THAL and IMIP treatment groups. These data paralleled the findings for aortic immunohistochemistry. The THAL group, but not the IMIP group, showed improved CMS–CCT-induced changes in the vascular reactivity of the aortic rings. Thus, TNF-α provides a target link between depression, metabolic syndrome, and endothelial dysfunction. This could open a new therapeutic approach to address the comorbidities of depression.
Collapse
Affiliation(s)
- Basma Ismail
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Abbassia, Cairo, Egypt
- Cardiac PET Centre, University of Ottawa Heart Institute, Ottawa, ON K1Y 4W7, Canada
| | - Sawsan Aboul-Fotouh
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Abbassia, Cairo, Egypt
- Clinical Pharmacology Unit, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Amal A. Mansour
- Department of Medical Biochemistry, Faculty of Medicine, Ain Shams University, Abbassia, Cairo, Egypt
| | - Hanan H. Shehata
- Department of Medical Biochemistry, Faculty of Medicine, Ain Shams University, Abbassia, Cairo, Egypt
| | - Manal I. Salman
- Department of Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Eman A. Ibrahim
- Department of Pathology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Olfat A. Hassan
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Abbassia, Cairo, Egypt
| | - Ahmed M. Abdel-tawab
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Abbassia, Cairo, Egypt
- Clinical Pharmacology Unit, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
21
|
Choi H, Nguyen HN, Lamb FS. Inhibition of endocytosis exacerbates TNF-α-induced endothelial dysfunction via enhanced JNK and p38 activation. Am J Physiol Heart Circ Physiol 2014; 306:H1154-63. [DOI: 10.1152/ajpheart.00885.2013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Tumor necrosis factor-α (TNF-α) is a pro-inflammatory cytokine that causes endothelial dysfunction. Endocytosis of TNF-α receptors (TNFR) precedes endosomal reactive oxygen species (ROS) production, which is required for NF-κB activation in vascular smooth muscle cells. It is unknown how endocytosis of TNFRs impacts signaling in endothelial cells. We hypothesized that TNF-α-induced endothelial dysfunction is induced by both endosomal and cell surface events, including NF-κB and mitogen-activated protein kinases (MAPKs) activation, and endocytosis of the TNFR modifies signaling. Mesenteric artery segments from C57BL/6 mice were treated with TNF-α (10 ng/ml) for 22 h in tissue culture, with or without signaling inhibitors (dynasore for endocytosis, SP600125 for JNK, SB203580 for p38, U0126 for ERK), and vascular function was assessed. Endothelium-dependent relaxation to acetylcholine (ACh) was impaired by TNF-α, and dynasore exacerbated this, whereas JNK or p38 inhibition prevented these effects. In cultured endothelial cells from murine mesenteric arteries, dynasore potentiated JNK and p38 but not ERK phosphorylation and promoted cell death. NF-κB activation by TNF-α was decreased by dynasore. JNK inhibition dramatically increased both the magnitude and duration of TNF-α-induced NF-κB activation and potentiated intercellular adhesion molecule-1 (ICAM-1) activation. Dynasore still inhibited NF-κB activation in the presence of SP600125. Thus TNF-α-induced endothelial dysfunction is both JNK and p38 dependent. Endocytosis modulates the balance of NF-κB and MAPK signaling, and inhibition of NF-κB activation by JNK limits this pro-proliferative signal, which may contribute to endothelial cell death in response to TNF-α.
Collapse
Affiliation(s)
- Hyehun Choi
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hong N. Nguyen
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Fred S. Lamb
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
22
|
Vaidyanathan K, Durning S, Wells L. Functional O-GlcNAc modifications: implications in molecular regulation and pathophysiology. Crit Rev Biochem Mol Biol 2014; 49:140-163. [PMID: 24524620 PMCID: PMC4912837 DOI: 10.3109/10409238.2014.884535] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
O-linked β-N-acetylglucosamine (O-GlcNAc) is a regulatory post-translational modification of intracellular proteins. The dynamic and inducible cycling of the modification is governed by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA) in response to UDP-GlcNAc levels in the hexosamine biosynthetic pathway (HBP). Due to its reliance on glucose flux and substrate availability, a major focus in the field has been on how O-GlcNAc contributes to metabolic disease. For years this post-translational modification has been known to modify thousands of proteins implicated in various disorders, but direct functional connections have until recently remained elusive. New research is beginning to reveal the specific mechanisms through which O-GlcNAc influences cell dynamics and disease pathology including clear examples of O-GlcNAc modification at a specific site on a given protein altering its biological functions. The following review intends to focus primarily on studies in the last half decade linking O-GlcNAc modification of proteins with chromatin-directed gene regulation, developmental processes, and several metabolically related disorders including Alzheimer's, heart disease and cancer. These studies illustrate the emerging importance of this post-translational modification in biological processes and multiple pathophysiologies.
Collapse
Affiliation(s)
| | - Sean Durning
- Complex Carbohydrate Research Center, University of Georgia, Athens, USA
| | - Lance Wells
- Complex Carbohydrate Research Center, University of Georgia, Athens, USA
| |
Collapse
|
23
|
Sen A, Most P, Peppel K. Induction of microRNA-138 by pro-inflammatory cytokines causes endothelial cell dysfunction. FEBS Lett 2014; 588:906-14. [PMID: 24486907 DOI: 10.1016/j.febslet.2014.01.033] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 01/17/2014] [Indexed: 01/25/2023]
Abstract
Exposure to pro-inflammatory cytokines, such as Angiotensin II, endothelin-1 or TNF leads to endothelial dysfunction, characterized by the reduced production of nitric oxide via endothelial nitric oxide synthase (eNOS). We recently identified the Ca(2+) binding protein S100A1 as an essential factor required for eNOS activity. Here we report that pro-inflammatory cytokines down-regulate expression of S100A1 in primary human microvascular endothelial cells (HMVECs) via induction of microRNA-138 (miR-138), in a manner that depends on the stabilization of HIF1-α. We show that loss of S100A1 in ECs reduces stimulus-induced NO production, which can be prevented by inhibition of miR-138. Our study suggests that targeting miR-138 might be beneficial for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Anagha Sen
- Center for Translational Medicine, Jefferson Medical College, Philadelphia, PA, USA
| | - Patrick Most
- Center for Translational Medicine, Jefferson Medical College, Philadelphia, PA, USA; Laboratory for Molecular and Translational Cardiology, Department of Internal Medicine III, University of Heidelberg, INF 350, 69120 Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg University Hospital, INF 410, 69120 Heidelberg, Germany
| | - Karsten Peppel
- Center for Translational Medicine, Jefferson Medical College, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Pregnancy Programming and Preeclampsia: Identifying a Human Endothelial Model to Study Pregnancy-Adapted Endothelial Function and Endothelial Adaptive Failure in Preeclamptic Subjects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 814:27-47. [DOI: 10.1007/978-1-4939-1031-1_4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
25
|
Hilgers RHP, Xing D, Gong K, Chen YF, Chatham JC, Oparil S. Acute O-GlcNAcylation prevents inflammation-induced vascular dysfunction. Am J Physiol Heart Circ Physiol 2012; 303:H513-22. [PMID: 22777418 DOI: 10.1152/ajpheart.01175.2011] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Acute increases in cellular protein O-linked N-acetyl-glucosamine (O-GlcNAc) modification (O-GlcNAcylation) have been shown to have protective effects in the heart and vasculature. We hypothesized that d-glucosamine (d-GlcN) and Thiamet-G, two agents that increase protein O-GlcNAcylation via different mechanisms, inhibit TNF-α-induced oxidative stress and vascular dysfunction by suppressing inducible nitric oxide (NO) synthase (iNOS) expression. Rat aortic rings were incubated for 3h at 37°C with d-GlcN or its osmotic control l-glucose (l-Glc) or with Thiamet-G or its vehicle control (H(2)O) followed by the addition of TNF-α or vehicle (H(2)O) for 21 h. After incubation, rings were mounted in a myograph to assess arterial reactivity. Twenty-four hours of incubation of aortic rings with TNF-α resulted in 1) a hypocontractility to 60 mM K(+) solution and phenylephrine, 2) blunted endothelium-dependent relaxation responses to ACh and substance P, and 3) unaltered relaxing response to the Ca(2+) ionophore A-23187 and the NO donor sodium nitroprusside compared with aortic rings cultured in the absence of TNF-α. d-GlcN and Thiamet-G pretreatment suppressed the TNF-α-induced hypocontractility and endothelial dysfunction. Total protein O-GlcNAc levels were significantly higher in aortic segments treated with d-GlcN or Thiamet-G compared with controls. Expression of iNOS protein was increased in TNF-α-treated rings, and this was attenuated by pretreatment with either d-GlcN or Thiamet-G. Dense immunostaining for nitrotyrosylated proteins was detected in the endothelium and media of the aortic wall, suggesting enhanced peroxynitrite production by iNOS. These findings demonstrate that acute increases in protein O-GlcNAcylation prevent TNF-α-induced vascular dysfunction, at least in part, via suppression of iNOS expression.
Collapse
Affiliation(s)
- Rob H P Hilgers
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama, Birmingham, Alabama 35294-0007, USA
| | | | | | | | | | | |
Collapse
|
26
|
Does anti-tnf therapy cause any change in platelet activation in ankylosing spondylitis patients? J Thromb Thrombolysis 2011; 33:154-9. [DOI: 10.1007/s11239-011-0663-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
27
|
Farb MG, Ganley-Leal L, Mott M, Liang Y, Ercan B, Widlansky ME, Bigornia SJ, Fiscale AJ, Apovian CM, Carmine B, Hess DT, Vita JA, Gokce N. Arteriolar function in visceral adipose tissue is impaired in human obesity. Arterioscler Thromb Vasc Biol 2011; 32:467-73. [PMID: 22095978 DOI: 10.1161/atvbaha.111.235846] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE The purpose of this study was to characterize the relationship between adipose tissue phenotype and depot-specific microvascular function in fat. METHODS AND RESULTS In 30 obese subjects (age 42±11 years, body mass index 46±11 kg/m(2)) undergoing bariatric surgery, we intraoperatively collected visceral and subcutaneous adipose tissue and characterized depot-specific adipose phenotypes. We assessed vasomotor function of the adipose microvasculature using videomicroscopy of small arterioles (75-250 μm) isolated from different fat compartments. Endothelium-dependent, acetylcholine-mediated vasodilation was severely impaired in visceral arterioles, compared to the subcutaneous depot (P<0.001 by ANOVA). Nonendothelium dependent responses to papaverine and nitroprusside were similar. Endothelial nitric oxide synthase inhibition with N(ω)-nitro-l-arginine methyl ester reduced subcutaneous vasodilation but had no effect on severely blunted visceral arteriolar responses. Visceral fat exhibited greater expression of proinflammatory, oxidative stress-related, hypoxia-induced, and proangiogenic genes; increased activated macrophage populations; and had a higher capacity for cytokine production ex vivo. CONCLUSIONS Our findings provide clinical evidence that the visceral microenvironment may be intrinsically toxic to arterial health providing a potential mechanism by which visceral adiposity burden is linked to atherosclerotic vascular disease. Our findings also support the evolving concept that both adipose tissue quality and quantity may play significant roles in shaping cardiovascular phenotypes in human obesity.
Collapse
Affiliation(s)
- Melissa G Farb
- Boston Medical Center, 88 East Newton St, D-8, Cardiology, Boston, MA 02118, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
El-Bassossy HM, El-Moselhy MA, Mahmoud MF. Pentoxifylline alleviates vascular impairment in insulin resistance via TNF-α inhibition. Naunyn Schmiedebergs Arch Pharmacol 2011; 384:277-85. [PMID: 21800096 DOI: 10.1007/s00210-011-0669-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 07/06/2011] [Indexed: 10/17/2022]
Abstract
Deterioration of vascular reactivity plays a pivotal role in vascular complications. Pentoxifylline (PTX) is a well-tolerated drug used to treat vascular insufficiency. We investigated the protective effect of PTX against vascular impairment in insulin resistance. Insulin resistance was induced by fructose (10%) in drinking water while PTX was concurrently administered (50 mg/kg(-1)) for 8 weeks. Serum levels of glucose, insulin, tumor necrosis factor alpha (TNF-α) were determined. Isolated aorta reactivity to phenylephrine (PE), potassium chloride (KCl), and acetylcholine (ACh) was studied, as was nitric oxide (NO) generation and histopathology. Insulin resistance was accompanied with a significant elevation in serum TNF-α level, marked leukocytes infiltration, and endothelial pyknosis. PTX inhibited insulin resistance and prevented TNF-α elevation, leukocyte infiltration and endothelial pyknosis. Vascular dysfunction was evident in insulin resistance as increased vascular contractility to PE and decreased relaxation to ACh, whereas PTX protected against this dysfunction. Notably, in vitro incubation with TNF-α (1 ng/ml(-1)) increased contractility to PE and decreased relaxation to ACh while concomitant PTX (1 mM) incubation partially restored response to ACh but not to PE. Furthermore, TNF-α reduced ACh-induced NO generation, whereeas PTX restored it. In conclusion, PTX protects from the impairment in vascular reactivity in insulin resistance, by a mechanism involving TNF-α inhibition.
Collapse
Affiliation(s)
- Hany M El-Bassossy
- Hypertension and Vascular Center, Wake Forest University Baptist Medical Center, Winston-Salem, NC, 27157, USA.
| | | | | |
Collapse
|
29
|
Félétou M. The Endothelium, Part I: Multiple Functions of the Endothelial Cells -- Focus on Endothelium-Derived Vasoactive Mediators. ACTA ACUST UNITED AC 2011. [DOI: 10.4199/c00031ed1v01y201105isp019] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
30
|
|
31
|
Abstract
Perivascular adipose tissue is a local deposit of adipose tissue surrounding the vasculature. Perivascular adipose tissue is present throughout the body and has been shown to have a local effect on blood vessels. The influence of perivascular adipose tissue on the vasculature changes with increasing adiposity. This article describes the anatomy and pathophysiology of perivascular adipose tissue and the experimental evidence supporting its local adverse effect on the vasculature. Methods for quantifying perivascular adipose tissue in free-living populations will be described. Finally, the epidemiological literature demonstrating an association between perivascular adipose tissue and cardiometabolic disease will be explored.
Collapse
Affiliation(s)
- Kathryn A Britton
- National Heart, Lung & Blood Institute’s Framingham Heart Study, Framingham, MA, USA
- Division of Cardiovascular Medicine, Brigham & Women’s Hospital & Harvard Medical School, Boston, MA, USA
| | - Caroline S Fox
- National Heart, Lung & Blood Institute’s Framingham Heart Study, Framingham, MA, USA
- National Heart, Lung & Blood Institute & the Center for Population Studies, Framingham, MA, USA
- Division of Endocrinology, Metabolism & Hypertension, Brigham & Women’s Hospital & Harvard Medical School, Boston, MA, USA
| |
Collapse
|
32
|
Zhang R, Ran HH, Gao YL, Ma J, Huang Y, Bai YG, Lin LJ. Differential vascular cell adhesion molecule-1 expression and superoxide production in simulated microgravity rat vasculature. EXCLI JOURNAL 2010; 9:195-204. [PMID: 29255400 PMCID: PMC5698884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Accepted: 12/22/2010] [Indexed: 10/30/2022]
Abstract
Exposure to microgravity leads to orthostatic intolerance in astronauts and differential vascular structural and functional adaptations have been implicated in its occurrence. The present study tended to clarify the characteristics of vascular inflammation and oxidative stress in hindlimb unweighting (HU) rat vasculature. Male Sprague-Dawley rats were randomly divided into control (CON) and hindlimb unweighting (HU) groups. Three weeks later, immunohistochemistry was used to localize the expression of vascular cell adhesion molecule-1 (VCAM-1) and laser scanning confocal microscope were used to detect superoxide production. Immunohistochemical results revealed positive staining of VCAM-1 on endothelial cells in HU rat basilar and carotid arteries compared with CON, but not in abdominal aorta and femoral arteries. Meanwhile, HU increased O2·- levels in all the layers of basilar and carotid arteries from HU rat but not in abdominal aorta and femoral arteries from HU rat. These data suggested that differential expression of VCAM-1 and O2·- production were concomitant with the vascular adaptations to simulated microgravity and whether they participate in vascular structure and function remodeling merits further investigation.
Collapse
Affiliation(s)
- Ran Zhang
- Institute of Geriatric Cardiology, Institute of Geriatric Cardiology, People's Liberation Army General Hospital, Beijing, 100853, China,*To whom correspondence should be addressed: Ran Zhang, Institute of Geriatric Cardiology, Institute of Geriatric Cardiology, People's Liberation Army General Hospital, Beijing, 100853, China, E-mail:
| | - Hai-hong Ran
- Department of Geriatric Hematology, Chinese People's Liberation Army General Hospital, Beijing, 100853, China
| | - Yu-ling Gao
- Institute of Geriatric Cardiology, Institute of Geriatric Cardiology, People's Liberation Army General Hospital, Beijing, 100853, China
| | - Jin Ma
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, 710032, China
| | - Ya Huang
- Institute of Geriatric Cardiology, Institute of Geriatric Cardiology, People's Liberation Army General Hospital, Beijing, 100853, China
| | - Yun-gang Bai
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, 710032, China
| | - Le-jian Lin
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
33
|
Abstract
The prevalence of obesity and diabetes is rising to epidemic proportions worldwide. Insulin resistance is central to the pathogenesis of type 2 diabetes. The role of insulin sensitive tissues and organs, such as adipose, liver, muscle and hypothalamus, in regulating insulin sensitivity has been extensively studied. However, the existence and nature of inter-tissue communication in the regulation of insulin action is unresolved. A major factor in the development of insulin resistance is obesity, especially abdominal obesity. Chronically positive energy balance leads to a process gradually progressing to insulin resistance in obese states. Increased adipocyte size is positively correlated with the frequency of adipocyte death in obesity. The death of the hypertrophic adipocytes facilitates the infiltration of macrophages, which further perpetuates adipose inflammation and insulin resistance, characterized by low liposynthetic capacity and high lipolytic capacity, causing increased release of free fatty acids (FFA). The inflamed adipose tissue releases FFA, inflammatory cytokines, and hormones that act as circulatory factors to regulate insulin sensitivity in distant organs. In addition, perivascular adipose tissue participates in regulating insulin sensitivity, vascular inflammation and function in an endocrine/paracrine manner through adipose-derived relaxing and contracting factors, cytokines and infiltration of inflammatory cells. Thus, adipose tissue releases cytokines, hormones and lipids that signal distant organs to regulate systemic metabolic homeostasis, as well as signal local blood vessels to modulate vascular inflammation and function. These findings may lead to uncovering new therapeutic targets to fight obesity and type 2 diabetes by restructuring the adipokine profile and lipid metabolism.
Collapse
Affiliation(s)
- Hanrui Zhang
- Department of Internal Medicine, University of Missouri-Columbia, Columbia, Missouri, USA.
| | | |
Collapse
|
34
|
Oxidative stress and inflammatory mediators contribute to endothelial dysfunction in high-fat diet-induced obesity in mice. J Hypertens 2010; 28:2111-9. [DOI: 10.1097/hjh.0b013e32833ca68c] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
35
|
El-Bassossy HM, El-Maraghy NN, El-Fayoumi HM, Watson ML. Haem oxygenase-1 induction protects against tumour necrosis factor alpha impairment of endothelial-dependent relaxation in rat isolated pulmonary artery. Br J Pharmacol 2009; 158:1527-35. [PMID: 19845678 DOI: 10.1111/j.1476-5381.2009.00419.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND AND PURPOSE Disturbances in pulmonary vascular reactivity are important components of inflammatory lung disease. Haem oxygenase-1 (HO-1) is an important homeostatic enzyme upregulated in inflammation. Here we have investigated the potentially protective effect of HO-1 against cytokine-induced impairment in pulmonary artery relaxation. EXPERIMENTAL APPROACH Haem oxygenase-1 protein levels were assessed by immunofluorescence. HO activity was assessed by conversion of haemin to bilirubin. Rings of rat isolated pulmonary artery in organ baths were used to measure relaxant responses to the endothelium-dependent agent ACh and the endothelium-independent agent sodium nitroprusside (SNP). Production of nitric oxide (NO) and reactive oxygen species (ROS) was assessed by confocal fluorescence microscopy and fluorescent probes. KEY RESULTS Haem oxygenase-1 protein expression was strongly induced in pulmonary artery after 24-h incubation with either haemin (5 microM) or curcumin (2 microM), accompanied by a significant increase in HO activity. Incubation with tumour necrosis factor alpha (TNFalpha, 1 ng.mL(-1), 2 h) significantly decreased relaxation of arterial rings to ACh, without affecting responses to SNP. Induction of HO-1 by curcumin or haemin protected against TNFalpha-induced hyporesponsiveness to ACh. The competitive HO inhibitor, tin protoporphyrin (20 microM), abolished the protective effect of haemin. HO-1 induction prevented a TNFalpha-induced increase in NO generation without affecting the TNFalpha-induced increase in ROS generation. HO-1 induction prevented the TNFalpha-induced decrease in ACh-stimulated NO generation. CONCLUSIONS AND IMPLICATIONS Induction of HO-1 protected against TNFalpha impairment of endothelium-dependent relaxation in pulmonary artery, by a mechanism involving a reduction in inducible NO synthase-derived NO production.
Collapse
|
36
|
Sprague AH, Khalil RA. Inflammatory cytokines in vascular dysfunction and vascular disease. Biochem Pharmacol 2009; 78:539-52. [PMID: 19413999 PMCID: PMC2730638 DOI: 10.1016/j.bcp.2009.04.029] [Citation(s) in RCA: 969] [Impact Index Per Article: 60.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2009] [Revised: 04/23/2009] [Accepted: 04/27/2009] [Indexed: 12/12/2022]
Abstract
The vascular inflammatory response involves complex interaction between inflammatory cells (neutrophils, lymphocytes, monocytes, macrophages), endothelial cells (ECs), vascular smooth muscle cells (VSMCs), and extracellular matrix (ECM). Vascular injury is associated with increased expression of adhesion molecules by ECs and recruitment of inflammatory cells, growth factors, and cytokines, with consequent effects on ECs, VSMCs and ECM. Cytokines include tumor necrosis factors, interleukins, lymphokines, monokines, interferons, colony stimulating factors, and transforming growth factors. Cytokines are produced by macrophages, T-cells and monocytes, as well as platelets, ECs and VSMCs. Circulating cytokines interact with specific receptors on various cell types and activate JAK-STAT, NF-kappaB, and Smad signaling pathways leading to an inflammatory response involving cell adhesion, permeability and apoptosis. Cytokines also interact with mitochondria to increase the production of reactive oxygen species. Cytokine-induced activation of these pathways in ECs modifies the production/activity of vasodilatory mediators such as nitric oxide, prostacyclin, endothelium-derived hyperpolarizing factor, and bradykinin, as well as vasoconstrictive mediators such as endothelin and angiotensin II. Cytokines interact with VSMCs to activate Ca(2+), protein kinase C, Rho-kinase, and MAPK pathways, which promote cell growth and migration, and VSM reactivity. Cytokines also interact with integrins and matrix metalloproteinases (MMPs) and modify ECM composition. Persistent increases in cytokines are associated with vascular dysfunction and vascular disease such as atherosclerosis, abdominal aortic aneurysm, varicose veins and hypertension. Genetic and pharmacological tools to decrease the production of cytokines or to diminish their effects using cytokine antagonists could provide new approaches in the management of inflammatory vascular disease.
Collapse
Affiliation(s)
- Alexander H Sprague
- Division of Vascular Surgery and Endovascular Therapy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
37
|
Greenstein AS, Khavandi K, Withers SB, Sonoyama K, Clancy O, Jeziorska M, Laing I, Yates AP, Pemberton PW, Malik RA, Heagerty AM. Local inflammation and hypoxia abolish the protective anticontractile properties of perivascular fat in obese patients. Circulation 2009; 119:1661-70. [PMID: 19289637 DOI: 10.1161/circulationaha.108.821181] [Citation(s) in RCA: 473] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Inflammation in adipose tissue has been implicated in vascular dysfunction, but the local mechanisms by which this occurs are unknown. METHODS AND RESULTS Small arteries with and without perivascular adipose tissue were taken from subcutaneous gluteal fat biopsy samples and studied with wire myography and immunohistochemistry. We established that healthy adipose tissue around human small arteries secretes factors that influence vasodilation by increasing nitric oxide bioavailability. However, in perivascular fat from obese subjects with metabolic syndrome (waist circumference 111+/-2.8 versus 91.1+/-3.5 cm in control subjects, P<0.001; insulin sensitivity 41+/-5.9% versus 121+/-18.6% in control subjects, P<0.001), the loss of this dilator effect was accompanied by an increase in adipocyte area (1786+/-346 versus 673+/-60 mum(2), P<0.01) and immunohistochemical evidence of inflammation (tumor necrosis factor receptor 1 12.4+/-1.1% versus 6.7+/-1%, P<0.001). Application of the cytokines tumor necrosis factor receptor-alpha and interleukin-6 to perivascular fat around healthy blood vessels reduced dilator activity, resulting in the obese phenotype. These effects could be reversed with free radical scavengers or cytokine antagonists. Similarly, induction of hypoxia stimulated inflammation and resulted in loss of anticontractile capacity, which could be rescued by catalase and superoxide dismutase or cytokine antagonists. Incubation with a soluble fragment of adiponectin type 1 receptor or inhibition of nitric oxide synthase blocked the vasodilator effect of healthy perivascular adipose tissue. CONCLUSIONS We conclude that adipocytes secrete adiponectin and provide the first functional evidence that it is a physiological modulator of local vascular tone by increasing nitric oxide bioavailability. This capacity is lost in obesity by the development of adipocyte hypertrophy, leading to hypoxia, inflammation, and oxidative stress.
Collapse
Affiliation(s)
- Adam S Greenstein
- Cardiovascular Research Group, University of Manchester, Manchester, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Healthy vascular function is primarily regulated by several factors including EDRF (endothelium-dependent relaxing factor), EDCF (endothelium-dependent contracting factor) and EDHF (endothelium-dependent hyperpolarizing factor). Vascular dysfunction or injury induced by aging, smoking, inflammation, trauma, hyperlipidaemia and hyperglycaemia are among a myriad of risk factors that may contribute to the pathogenesis of many cardiovascular diseases, such as hypertension, diabetes and atherosclerosis. However, the exact mechanisms underlying the impaired vascular activity remain unresolved and there is no current scientific consensus. Accumulating evidence suggests that the inflammatory cytokine TNF (tumour necrosis factor)-α plays a pivotal role in the disruption of macrovascular and microvascular circulation both in vivo and in vitro. AGEs (advanced glycation end-products)/RAGE (receptor for AGEs), LOX-1 [lectin-like oxidized low-density lipoprotein receptor-1) and NF-κB (nuclear factor κB) signalling play key roles in TNF-α expression through an increase in circulating and/or local vascular TNF-α production. The increase in TNF-α expression induces the production of ROS (reactive oxygen species), resulting in endothelial dysfunction in many pathophysiological conditions. Lipid metabolism, dietary supplements and physical activity affect TNF-α expression. The interaction between TNF-α and stem cells is also important in terms of vascular repair or regeneration. Careful scrutiny of these factors may help elucidate the mechanisms that induce vascular dysfunction. The focus of the present review is to summarize recent evidence showing the role of TNF-α in vascular dysfunction in cardiovascular disease. We believe these findings may prompt new directions for targeting inflammation in future therapies.
Collapse
|
39
|
Bonaventura D, Tirapelli CR, de Oliveira AM. Chronic methionine load-induced hyperhomocysteinemia impairs the relaxation induced by bradykinin in the isolated rat carotid. Amino Acids 2008; 37:617-27. [DOI: 10.1007/s00726-008-0181-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Accepted: 09/06/2008] [Indexed: 11/29/2022]
|
40
|
Gillham JC, Myers JE, Baker PN, Taggart MJ. TNF-alpha alters nitric oxide- and endothelium-derived hyperpolarizing factor-mediated vasodilatation in human omental arteries. Hypertens Pregnancy 2008; 27:29-38. [PMID: 18293202 DOI: 10.1080/10641950701825796] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To investigate the effect of TNF-alpha on the endothelial function of human systemic arteries. METHODS Omental arteries were obtained from healthy pregnant women undergoing Cesarean section and examined using isometric wire myography. RESULTS Incubation with TNF-alpha (1nM) alone did not alter bradykinin-mediated endothelium-dependent relaxation of arteries. However, TNF-alpha did attenuate nitric oxide- (NO) and prostacyclin-independent endothelial-mediated relaxation. Similarly, in vessels constricted with a high potassium solution (60 mM), which inhibits vasodilatation via endothelial-derived hyperpolarising factor (EDHF), TNF-alpha incubation also attenuated bradykinin-induced vasodilatation. CONCLUSIONS The vasorelaxant capacity of human systemic arteries is compromised by TNF-alpha incubation in the presence of NO/prostacyclin or EDHF-blockade.
Collapse
Affiliation(s)
- J C Gillham
- Maternal and Fetal Health Research Centre, Division of Human Development, St Mary's Hospital, Manchester, UK
| | | | | | | |
Collapse
|
41
|
Zhang R, Jia G, Bao J, Zhang Y, Bai Y, Lin L, Tang H, Ma J. Increased Vascular Cell Adhesion Molecule–1 Was Associated with Impaired Endothelium–Dependent Relaxation of Cerebral and Carotid Arteries in Simulated Microgravity Rats. J Physiol Sci 2008; 58:67-73. [DOI: 10.2170/physiolsci.rp010707] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Accepted: 01/28/2008] [Indexed: 11/05/2022]
|
42
|
Tirapelli CR, Bonaventura D, de Oliveira AM. Functional characterization of the mechanisms underlying bradykinin-induced relaxation in the isolated rat carotid artery. Life Sci 2007; 80:1799-805. [PMID: 17367816 DOI: 10.1016/j.lfs.2007.02.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Accepted: 02/07/2007] [Indexed: 10/23/2022]
Abstract
This work aimed to functionally characterize the mechanisms underlying the relaxation induced by bradykinin (BK) in the rat carotid artery. Vascular reactivity experiments, using standard muscle bath procedures, showed that BK (0.1 nmol/L-3 mumol/L) induced relaxation of phenylephrine-pre-contracted rings in a concentration-dependent manner. Endothelial removal strongly attenuated BK-induced relaxation. HOE-140, the selective antagonist of bradykinin B(2) receptors concentration-dependently reduced the relaxation induced by BK. Pre-incubation of endothelium-intact rings with L-NAME (100 micromol/L), a non-selective nitric oxide synthase (NOS) inhibitor, L-NAME (100 micromol/L), a selective inhibitor of the eNOS or 7-nitroindazole (100 micromol/L), the selective inhibitor of nNOS, reduced BK-induced relaxation. Conversely, 1400 W (10 nmol/L), a selective inhibitor of iNOS, did not alter the relaxation induced by BK. Surprisingly, indomethacin (10 micromol/L) a non-selective inhibitor of cyclooxygenase (COX) increased BK-induced relaxation in endothelium-intact but not denuded rings. Neither SQ29548 (3 micromol/L), a competitive antagonist of PGH(2)/TXA(2) receptors nor AH6809 (10 micromol/L), an antagonist of PGF(2alpha) receptors significantly altered the relaxation induced by BK in endothelium-intact rings. The combination of SQ29548 and AH6809 increased BK-induced relaxation. The present study shows that the vasorelaxant action displayed by BK in the rat carotid is mediated by endothelial B(2) receptors and the activation of the NO pathway. The major finding of this work is that it demonstrated functionally that endothelial-derived vasoconstrictor prostanoids (probably PGH(2), TXA(2) and PGF(2alpha)) counteract the vasorelaxant action displayed by BK.
Collapse
Affiliation(s)
- Carlos R Tirapelli
- Department of Psychiatric Nursing and Human Sciences, College of Nursing of Ribeirão Preto, University of São Paulo (USP), SP, Brazil
| | | | | |
Collapse
|
43
|
Endothelial nitric oxide synthase in human intestine resected for necrotizing enterocolitis. J Pediatr 2007; 150:40-5. [PMID: 17188611 DOI: 10.1016/j.jpeds.2006.09.029] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2006] [Revised: 07/18/2006] [Accepted: 09/01/2006] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To determine the expression and function of endothelial nitric oxide synthase (eNOS) in submucosal arterioles harvested from human intestine resected for necrotizing enterocolitis (NEC) or congenital bowel disease. STUDY DESIGN eNOS expression was determined by using immunohistochemistry. The arteriolar diameter was measured in vitro at pressures of 10 to 40 mm Hg and also in response to the eNOS agonist acetylcholine (ACh), the exogenous nitric oxide (NO) donor S-nitroso-N-acetylpenicillamine, and the smooth muscle relaxant papaverine. Arteriolar release of NO in response to ACh was determined with a Sievers NOAnalyzer. Hemodynamics were also determined at flow rates of 50 and 100 microL/min. RESULTS eNOS was present in microvessels from both groups, but NEC arterioles failed to demonstrate physiological evidence of eNOS function: they constricted in response to pressure, failed to dilate or generate NO in response to ACh, and failed to dilate in response to flow. However, they dilated in response to exogenous NO and papaverine, indicating functional vascular smooth muscle and vasodilator reserve. CONCLUSION eNOS-derived NO, a vasodilator in the newborn intestine, did not contribute to vasoregulation in arterioles harvested from intestine resected for NEC. These vessels were constricted; lack of eNOS-derived NO may contribute to this vasoconstriction.
Collapse
|
44
|
Abstract
Studies were carried out to determine the effects of IL-1beta on newborn intestinal hemodynamics. IL-1beta increased the release of ET-1 by primary endothelial cells in a dose-dependent manner; as well, it reduced expression of the endothelin (ET) type B (ET(B)) receptor on endothelial cells and increased expression of the ET type A (ET(A)) receptor on vascular smooth muscle cells. IL-1beta increased endothelial cell endothelial nitric oxide (NO) synthase (eNOS) expression but did not enhance eNOS activity as evidenced by release of NO(x) into conditioned medium in response to acetylcholine or shear stress. The effects of IL-1beta on flow-induced dilation were evaluated in terminal mesenteric arteries in vitro. Pretreatment with IL-1beta (1 ng; 4 h) significantly attenuated vasodilation in response to flow rates of 100 and 200 microl/min. This effect was mediated, in part, by the endothelin ET(A) receptor; thus selective blockade of ET(A) receptors with BQ610 nearly restored flow-induced dilation. In contrast, exogenous ET-1 only shifted the diameter-flow curve downward without altering the percent vasodilation in response to flow. The effects of IL-1beta on ileal oxygenation were then studied using in vivo gut loops. Intramesenteric artery infusion of IL-1beta upstream of the gut loop caused ileal vasoconstriction and reduced the arterial-venous O(2) difference across the gut loop; consequently, it reduced ileal oxygenation by 60%. This effect was significantly attenuated by pretreatment with BQ610. These data support a linkage between the proinflammatory cytokine IL-1beta and vascular dysfunction within the intestinal circulation, mediated, at least in part, by the ET system.
Collapse
Affiliation(s)
- Philip T Nowicki
- Columbus Children's Research Institute, Children's Hospital, 700 Children's Dr., Columbus, OH 43205, USA.
| |
Collapse
|
45
|
Raza K, Carruthers DM, Stevens R, Filer AD, Townend JN, Bacon PA. Infliximab leads to a rapid but transient improvement in endothelial function in patients with primary systemic vasculitis. Ann Rheum Dis 2006; 65:946-8. [PMID: 16361277 PMCID: PMC1798195 DOI: 10.1136/ard.2005.043638] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2005] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To assess the immediate effects of tumour necrosis factor alpha (TNFalpha) blockade on endothelial function in systemic vasculitis. METHODS Endothelial function was assessed by laser Doppler flowmetry in patients with active vasculitis after 10 infusions of infliximab. For comparison endothelial responses were assessed after five infusions of cyclophosphamide plus methylprednisolone. RESULTS Endothelial dependent vasodilatation (EDV) improved significantly within 24 hours of infliximab infusion. The median change in red blood cell flux (interquartile range) was 5.7 (4.3-8.2) before infusion v 8.4 (7.5-10.9) at 24 hours; p=0.027. This was not maintained at day 14. No improvement was seen in EDV after cyclophosphamide plus methylprednisolone infusion. CONCLUSION The rapid but transient improvement in EDV after TNFalpha inhibition suggests that TNFalpha may have a direct role in the impairment of endothelial function.
Collapse
Affiliation(s)
- K Raza
- Rheumatology Research Group, Division of Immunity and Infection, The Medical School, The University of Birmingham, and Department of Rheumatology, City Hospital, UK
| | | | | | | | | | | |
Collapse
|
46
|
Jiménez-Altayó F, Briones AM, Giraldo J, Planas AM, Salaices M, Vila E. Increased superoxide anion production by interleukin-1beta impairs nitric oxide-mediated relaxation in resistance arteries. J Pharmacol Exp Ther 2006; 316:42-52. [PMID: 16183707 DOI: 10.1124/jpet.105.088435] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The present study was designed to analyze the effect of long-term incubation with interleukin-1beta (IL-1beta) on endothelium-dependent relaxation in rat mesenteric resistance arteries. Vessels were incubated in culture medium with or without IL-1beta (10 ng/ml, 14 h). Changes in lumen diameter were recorded in a pressure myograph. Protein expression, nitrite, and superoxide anion (O(2)(.)) production were evaluated by either Western blot or immunofluorescence, Griess reaction, and ethidium fluorescence, respectively. IL-1beta impaired acetylcholine (ACh) and sodium nitroprusside (SNP) vasodilation and increased nitrite and O(2)(.) production and inducible nitric-oxide synthase (iNOS), xanthine oxidase, and p22(phox) expression. However, neither endothelial nitric-oxide synthase (NOS) nor soluble guanylate cyclase protein expression were affected by IL-1beta treatment. Polyethylene glycol superoxide dismutase partially reversed the impairment of ACh relaxation and abolished the O(2)(.) production observed in IL-1beta-treated arteries. The impairment of ACh relaxation induced by IL-1beta was also partially reversed by the xanthine oxidase inhibitor allopurinol (1 mM) but not by either the NADPH oxidase inhibitor apocynin (0.3 mM) or the inducible NOS inhibitor N-3-aminomethylbenzylacetamidine (1 microM). However, all these inhibitors improved the impaired SNP response. The results of the present study demonstrate that long-term incubation with IL-1beta induces an impairment of the nitric oxide-mediated relaxation in mesenteric resistance arteries through the production of O(2)(.), mainly from xanthine oxidase.
Collapse
Affiliation(s)
- Francesc Jiménez-Altayó
- Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | | | | | | | | | | |
Collapse
|
47
|
Cheranov SY, Jaggar JH. TNF-alpha dilates cerebral arteries via NAD(P)H oxidase-dependent Ca2+ spark activation. Am J Physiol Cell Physiol 2005; 290:C964-71. [PMID: 16267103 PMCID: PMC1638900 DOI: 10.1152/ajpcell.00499.2005] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Expression of TNF-alpha, a pleiotropic cytokine, is elevated during stroke and cerebral ischemia. TNF-alpha regulates arterial diameter, although mechanisms mediating this effect are unclear. In the present study, we tested the hypothesis that TNF-alpha regulates the diameter of resistance-sized ( approximately 150-microm diameter) cerebral arteries by modulating local and global intracellular Ca(2+) signals in smooth muscle cells. Laser-scanning confocal imaging revealed that TNF-alpha increased Ca(2+) spark and Ca(2+) wave frequency but reduced global intracellular Ca(2+) concentration ([Ca(2+)](i)) in smooth muscle cells of intact arteries. TNF-alpha elevated reactive oxygen species (ROS) in smooth muscle cells of intact arteries, and this increase was prevented by apocynin or diphenyleneiodonium (DPI), both of which are NAD(P)H oxidase blockers, but was unaffected by inhibitors of other ROS-generating enzymes. In voltage-clamped (-40 mV) cells, TNF-alpha increased the frequency and amplitude of Ca(2+) spark-induced, large-conductance, Ca(2+)-activated K(+) (K(Ca)) channel transients approximately 1.7- and approximately 1.4-fold, respectively. TNF-alpha-induced transient K(Ca) current activation was reversed by apocynin or by Mn(III)tetrakis(1-methyl-4-pyridyl)porphyrin (MnTMPyP), a membrane-permeant antioxidant, and was prevented by intracellular dialysis of catalase. TNF-alpha induced reversible and similar amplitude dilations in either endothelium-intact or endothelium-denuded pressurized (60 mmHg) cerebral arteries. MnTMPyP, thapsigargin, a sarcoplasmic reticulum Ca(2+)-ATPase blocker that inhibits Ca(2+) sparks, and iberiotoxin, a K(Ca) channel blocker, reduced TNF-alpha-induced vasodilations to between 15 and 33% of control. In summary, our data indicate that TNF-alpha activates NAD(P)H oxidase, resulting in an increase in intracellular H(2)O(2) that stimulates Ca(2+) sparks and transient K(Ca) currents, leading to a reduction in global [Ca(2+)](i), and vasodilation.
Collapse
Affiliation(s)
- Sergey Y Cheranov
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, 426 Nash Research Bldg., Memphis, 38163, USA
| | | |
Collapse
|
48
|
Arenas IA, Armstrong SJ, Xu Y, Davidge ST. Chronic tumor necrosis factor-alpha inhibition enhances NO modulation of vascular function in estrogen-deficient rats. Hypertension 2005; 46:76-81. [PMID: 15911738 DOI: 10.1161/01.hyp.0000168925.98963.ef] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tumor necrosis factor-alpha (TNF-alpha) is involved in the pathogenesis of vascular disease. Clinical studies have shown that postmenopausal women have higher serum TNF-alpha levels; however, whether this increase in TNF-alpha is associated with vascular dysfunction is unknown. We investigated whether estrogen deficiency is associated with increased serum TNF-alpha levels and tested the effects of in vivo TNF-alpha inhibition on vascular reactivity. Aged (12 to 15 months) Sprague-Dawley rats were ovariectomized and treated with placebo, estrogen, or a TNF-alpha inhibitor (Etanercept; 0.3 mg/kg) for 4 weeks. Serum TNF-alpha was determined by a bioassay, and vascular function was evaluated in the myograph system. Estrogen-deficient animals had higher serum levels of TNF-alpha compared with either estrogen-replaced animals or animals treated with Etanercept. Moreover, in estrogen-deficient rats, TNF-alpha inhibition reduced the constriction of mesenteric arteries to phenylephrine, increased the modulation of this vasoconstriction by the NO synthase inhibitor nitro-l-arginine methyl ester, and decreased the modulation by a superoxide scavenger (Mn(III)tetrakis(4-benzoic acid) porphyrin chloride). Furthermore, endothelium-dependent relaxation was also enhanced by TNF-alpha antagonism. Additionally, vascular expression of endothelial NO synthase was increased in animals treated with Etanercept, whereas the expression of NAD(P)H oxidase gp91phox and p22phox subunits was decreased. These data show that estrogen-deficient female rats have higher bioactive serum TNF-alpha levels compared with estrogen-replaced animals. Moreover, a decrease in serum bioactive TNF-alpha by a soluble TNF-alpha receptor (Etanercept) results in increased modulation of vascular function by NO. These observations suggest that TNF-alpha could be a mediator of vascular dysfunction associated with estrogen deficiency.
Collapse
Affiliation(s)
- Ivan A Arenas
- Perinatal Research Center, Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| | | | | | | |
Collapse
|
49
|
Abstract
Cytokine levels are elevated in many cardiovascular diseases and seem to be implicated in the associated disturbances in vascular reactivity reported in these diseases. Arterial blood pressure is maintained within a normal range by changes in peripheral resistance and cardiac output. Peripheral resistance is mainly determined by small resistance arteries and arterioles. This review focuses on the effects of cytokines, mainly TNF-α, IL-1β, and IL-6, on the reactivity of resistance arteries. The vascular effects of cytokines depend on the balance between the vasoactive mediators released under their influence in the different vascular beds. Cytokines may induce a vasodilatation and hyporesponsiveness to vasoconstrictors that may be relevant to the pathogenesis of septic shock. Cytokines may also induce vasoconstriction or increase the response to vasoconstrictor agents and impair endothelium-dependent vasodilatation. These effects may help predispose to vessel spasm, thrombosis, and atherogenesis and reinforce the link between inflammation and vascular disease.
Collapse
|
50
|
Abstract
One of the most important questions in vasculitis research is not why inflammation of blood vessels occurs but why it persists, often in a site-specific manner. In this review we illustrate how stromal cells, such as fibroblasts and pericytes, might play an important role in regulating the site at which vasculitis occurs. Smooth muscle cells and fibroblasts directly influence the behaviour of overlying vascular cells, amplifying the response of the endothelium to proinflammatory agents such as TNF-alpha and allowing enhanced and inappropriate leucocyte recruitment. An abnormal local vascular stromal environment can therefore influence local endothelial function and drive the persistence of local vascular inflammation. However, such local vascular inflammation can have distant effects on the systemic vascular system, leading to widespread endothelial cell dysfunction. Vascular endothelial dysfunction is common in a range of immune-mediated inflammatory diseases, is seen in multiple vascular beds, and is reversible following the induction of disease remission. The mechanisms that drive such systemic vascular endothelial dysfunction are unclear but factors such as TNF-alpha and CRP may play a role. Persistence of such widespread endothelial dysfunction in systemic vasculitis appears to have long-term consequences, leading to the acceleration of atherosclerosis and premature ischaemic heart disease. It may also underlie the accelerated atherosclerosis seen in other immune-mediated rheumatic diseases, such as rheumatoid arthritis.
Collapse
Affiliation(s)
- Christopher D Buckley
- Department of Rheumatology, Division of Immunity and Infection, University of Birmingham, Birmingham B15 2TT, UK.
| | | | | | | |
Collapse
|