1
|
de Oliveira HT, Couto GK, Davel AP, Xavier FE, Rossoni LV. Chronic cyclooxygenase-2 inhibition prevents the worsening of hypertension and endothelial dysfunction induced by ouabain in resistance arteries of spontaneously hypertensive rats. Vascul Pharmacol 2021; 139:106880. [PMID: 34052431 DOI: 10.1016/j.vph.2021.106880] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/21/2021] [Accepted: 05/25/2021] [Indexed: 01/16/2023]
Abstract
AIM Previous studies raise cyclooxygenase (COX) activation as a possible mechanism involved in the pathophysiology of ouabain-induced hypertension. We hypothesized that inhibition of COX-2 activity might prevent ouabain-induced vascular dysfunction and worsening of hypertension in spontaneously hypertensive rats (SHR). METHODS SHR were exposed to ouabain or vehicle and treated or not with the selective COX-2 inhibitor nimesulide for 5 weeks. Systolic blood pressure was measured by plethysmography. Vascular reactivity by wire myograph and protein expression by Western-blot were assessed in mesenteric resistance arteries (MRA) of groups. Thromboxane A2 (TXA2) production by ELISA was evaluated in MRA supernatants of groups. RESULTS Noradrenaline-induced maximal contraction (Emax) was greater in MRA from SHR receiving ouabain than those of vehicle group. In situ inhibition of COX-2, TXA2 synthase, or TP receptor reduced the Emax to noradrenaline in MRA of ouabain to vehicle levels. TXA2 production was higher in ouabain than in vehicle group. Ouabain enhanced expression of cytoplasmic tyrosine kinase Src (c-Src)/ERK1/2/COX-2/TXA2 synthase/TP receptor in SHR MRA, but did not change NFkB/iKB ratio. Anticontractile effect of nitric oxide (NO) was smaller in MRA from ouabain- than vehicle-treated SHR, as well as eNOS and nNOS expression. Nimesulide co-treatment prevented the ouabain-induced worsening of hypertension and noradrenaline MRA hypercontractility in SHR. CONCLUSION Ouabain worsen hypertension and induce MRA hypercontractility in SHR associated with upregulated c-Src/ERK1/2/COX-2/TXA2 synthase/TXA2/TP receptor axis. These effects were prevented by COX-2 inhibition.
Collapse
Affiliation(s)
- Helane Tito de Oliveira
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Gisele Kruger Couto
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, Brazil
| | - Ana Paula Davel
- Department of Structural and Functional Biology, University of Campinas, Campinas, Brazil
| | - Fabiano Elias Xavier
- Department of Physiology and Pharmacology, Biosciences Center, Federal University of Pernambuco, Recife, Brazil
| | - Luciana Venturini Rossoni
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
2
|
Boguslavskyi A, Tokar S, Prysyazhna O, Rudyk O, Sanchez-Tatay D, Lemmey HA, Dora KA, Garland CJ, Warren HR, Doney A, Palmer CN, Caulfield MJ, Vlachaki Walker J, Howie J, Fuller W, Shattock MJ. Phospholemman Phosphorylation Regulates Vascular Tone, Blood Pressure, and Hypertension in Mice and Humans. Circulation 2021; 143:1123-1138. [PMID: 33334125 PMCID: PMC7969167 DOI: 10.1161/circulationaha.119.040557] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 12/09/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND Although it has long been recognized that smooth muscle Na/K ATPase modulates vascular tone and blood pressure (BP), the role of its accessory protein phospholemman has not been characterized. The aim of this study was to test the hypothesis that phospholemman phosphorylation regulates vascular tone in vitro and that this mechanism plays an important role in modulation of vascular function and BP in experimental models in vivo and in humans. METHODS In mouse studies, phospholemman knock-in mice (PLM3SA; phospholemman [FXYD1] in which the 3 phosphorylation sites on serines 63, 68, and 69 are mutated to alanines), in which phospholemman is rendered unphosphorylatable, were used to assess the role of phospholemman phosphorylation in vitro in aortic and mesenteric vessels using wire myography and membrane potential measurements. In vivo BP and regional blood flow were assessed using Doppler flow and telemetry in young (14-16 weeks) and old (57-60 weeks) wild-type and transgenic mice. In human studies, we searched human genomic databases for mutations in phospholemman in the region of the phosphorylation sites and performed analyses within 2 human data cohorts (UK Biobank and GoDARTS [Genetics of Diabetes Audit and Research in Tayside]) to assess the impact of an identified single nucleotide polymorphism on BP. This single nucleotide polymorphism was expressed in human embryonic kidney cells, and its effect on phospholemman phosphorylation was determined using Western blotting. RESULTS Phospholemman phosphorylation at Ser63 and Ser68 limited vascular constriction in response to phenylephrine. This effect was blocked by ouabain. Prevention of phospholemman phosphorylation in the PLM3SA mouse profoundly enhanced vascular responses to phenylephrine both in vitro and in vivo. In aging wild-type mice, phospholemman was hypophosphorylated, and this correlated with the development of aging-induced essential hypertension. In humans, we identified a nonsynonymous coding variant, single nucleotide polymorphism rs61753924, which causes the substitution R70C in phospholemman. In human embryonic kidney cells, the R70C mutation prevented phospholemman phosphorylation at Ser68. This variant's rare allele is significantly associated with increased BP in middle-aged men. CONCLUSIONS These studies demonstrate the importance of phospholemman phosphorylation in the regulation of vascular tone and BP and suggest a novel mechanism, and therapeutic target, for aging-induced essential hypertension in humans.
Collapse
Affiliation(s)
- Andrii Boguslavskyi
- British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.B., S.T., O.P., O.R., D.S.-T., M.J.S.). Clinical Pharmacology, The William Harvey Research Institute (O.P., H.R.W., M.J.C.), National Institute for Health Research, Biomedical Research Centre (H.R.W., M.J.C.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom. Department of Pharmacology, University of Oxford, United Kingdom (H.A.L.L., K.A.D., C.J.G.). Medicines Monitoring Unit, School of Medicine (A.D.), Division of Cardiovascular and Diabetes Medicine (C.N.A.), University of Dundee, United Kingdom. Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (J.V.W., J.H., W.F.)
| | - Sergiy Tokar
- British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.B., S.T., O.P., O.R., D.S.-T., M.J.S.). Clinical Pharmacology, The William Harvey Research Institute (O.P., H.R.W., M.J.C.), National Institute for Health Research, Biomedical Research Centre (H.R.W., M.J.C.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom. Department of Pharmacology, University of Oxford, United Kingdom (H.A.L.L., K.A.D., C.J.G.). Medicines Monitoring Unit, School of Medicine (A.D.), Division of Cardiovascular and Diabetes Medicine (C.N.A.), University of Dundee, United Kingdom. Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (J.V.W., J.H., W.F.)
| | - Oleksandra Prysyazhna
- British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.B., S.T., O.P., O.R., D.S.-T., M.J.S.). Clinical Pharmacology, The William Harvey Research Institute (O.P., H.R.W., M.J.C.), National Institute for Health Research, Biomedical Research Centre (H.R.W., M.J.C.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom. Department of Pharmacology, University of Oxford, United Kingdom (H.A.L.L., K.A.D., C.J.G.). Medicines Monitoring Unit, School of Medicine (A.D.), Division of Cardiovascular and Diabetes Medicine (C.N.A.), University of Dundee, United Kingdom. Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (J.V.W., J.H., W.F.)
| | - Olena Rudyk
- British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.B., S.T., O.P., O.R., D.S.-T., M.J.S.). Clinical Pharmacology, The William Harvey Research Institute (O.P., H.R.W., M.J.C.), National Institute for Health Research, Biomedical Research Centre (H.R.W., M.J.C.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom. Department of Pharmacology, University of Oxford, United Kingdom (H.A.L.L., K.A.D., C.J.G.). Medicines Monitoring Unit, School of Medicine (A.D.), Division of Cardiovascular and Diabetes Medicine (C.N.A.), University of Dundee, United Kingdom. Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (J.V.W., J.H., W.F.)
| | - David Sanchez-Tatay
- British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.B., S.T., O.P., O.R., D.S.-T., M.J.S.). Clinical Pharmacology, The William Harvey Research Institute (O.P., H.R.W., M.J.C.), National Institute for Health Research, Biomedical Research Centre (H.R.W., M.J.C.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom. Department of Pharmacology, University of Oxford, United Kingdom (H.A.L.L., K.A.D., C.J.G.). Medicines Monitoring Unit, School of Medicine (A.D.), Division of Cardiovascular and Diabetes Medicine (C.N.A.), University of Dundee, United Kingdom. Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (J.V.W., J.H., W.F.)
| | - Hamish A.L. Lemmey
- British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.B., S.T., O.P., O.R., D.S.-T., M.J.S.). Clinical Pharmacology, The William Harvey Research Institute (O.P., H.R.W., M.J.C.), National Institute for Health Research, Biomedical Research Centre (H.R.W., M.J.C.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom. Department of Pharmacology, University of Oxford, United Kingdom (H.A.L.L., K.A.D., C.J.G.). Medicines Monitoring Unit, School of Medicine (A.D.), Division of Cardiovascular and Diabetes Medicine (C.N.A.), University of Dundee, United Kingdom. Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (J.V.W., J.H., W.F.)
| | - Kim A. Dora
- British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.B., S.T., O.P., O.R., D.S.-T., M.J.S.). Clinical Pharmacology, The William Harvey Research Institute (O.P., H.R.W., M.J.C.), National Institute for Health Research, Biomedical Research Centre (H.R.W., M.J.C.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom. Department of Pharmacology, University of Oxford, United Kingdom (H.A.L.L., K.A.D., C.J.G.). Medicines Monitoring Unit, School of Medicine (A.D.), Division of Cardiovascular and Diabetes Medicine (C.N.A.), University of Dundee, United Kingdom. Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (J.V.W., J.H., W.F.)
| | - Christopher J. Garland
- British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.B., S.T., O.P., O.R., D.S.-T., M.J.S.). Clinical Pharmacology, The William Harvey Research Institute (O.P., H.R.W., M.J.C.), National Institute for Health Research, Biomedical Research Centre (H.R.W., M.J.C.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom. Department of Pharmacology, University of Oxford, United Kingdom (H.A.L.L., K.A.D., C.J.G.). Medicines Monitoring Unit, School of Medicine (A.D.), Division of Cardiovascular and Diabetes Medicine (C.N.A.), University of Dundee, United Kingdom. Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (J.V.W., J.H., W.F.)
| | - Helen R. Warren
- British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.B., S.T., O.P., O.R., D.S.-T., M.J.S.). Clinical Pharmacology, The William Harvey Research Institute (O.P., H.R.W., M.J.C.), National Institute for Health Research, Biomedical Research Centre (H.R.W., M.J.C.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom. Department of Pharmacology, University of Oxford, United Kingdom (H.A.L.L., K.A.D., C.J.G.). Medicines Monitoring Unit, School of Medicine (A.D.), Division of Cardiovascular and Diabetes Medicine (C.N.A.), University of Dundee, United Kingdom. Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (J.V.W., J.H., W.F.)
| | - Alexander Doney
- British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.B., S.T., O.P., O.R., D.S.-T., M.J.S.). Clinical Pharmacology, The William Harvey Research Institute (O.P., H.R.W., M.J.C.), National Institute for Health Research, Biomedical Research Centre (H.R.W., M.J.C.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom. Department of Pharmacology, University of Oxford, United Kingdom (H.A.L.L., K.A.D., C.J.G.). Medicines Monitoring Unit, School of Medicine (A.D.), Division of Cardiovascular and Diabetes Medicine (C.N.A.), University of Dundee, United Kingdom. Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (J.V.W., J.H., W.F.)
| | - Colin N.A. Palmer
- British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.B., S.T., O.P., O.R., D.S.-T., M.J.S.). Clinical Pharmacology, The William Harvey Research Institute (O.P., H.R.W., M.J.C.), National Institute for Health Research, Biomedical Research Centre (H.R.W., M.J.C.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom. Department of Pharmacology, University of Oxford, United Kingdom (H.A.L.L., K.A.D., C.J.G.). Medicines Monitoring Unit, School of Medicine (A.D.), Division of Cardiovascular and Diabetes Medicine (C.N.A.), University of Dundee, United Kingdom. Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (J.V.W., J.H., W.F.)
| | - Mark J. Caulfield
- British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.B., S.T., O.P., O.R., D.S.-T., M.J.S.). Clinical Pharmacology, The William Harvey Research Institute (O.P., H.R.W., M.J.C.), National Institute for Health Research, Biomedical Research Centre (H.R.W., M.J.C.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom. Department of Pharmacology, University of Oxford, United Kingdom (H.A.L.L., K.A.D., C.J.G.). Medicines Monitoring Unit, School of Medicine (A.D.), Division of Cardiovascular and Diabetes Medicine (C.N.A.), University of Dundee, United Kingdom. Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (J.V.W., J.H., W.F.)
| | - Julia Vlachaki Walker
- British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.B., S.T., O.P., O.R., D.S.-T., M.J.S.). Clinical Pharmacology, The William Harvey Research Institute (O.P., H.R.W., M.J.C.), National Institute for Health Research, Biomedical Research Centre (H.R.W., M.J.C.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom. Department of Pharmacology, University of Oxford, United Kingdom (H.A.L.L., K.A.D., C.J.G.). Medicines Monitoring Unit, School of Medicine (A.D.), Division of Cardiovascular and Diabetes Medicine (C.N.A.), University of Dundee, United Kingdom. Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (J.V.W., J.H., W.F.)
| | - Jacqueline Howie
- British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.B., S.T., O.P., O.R., D.S.-T., M.J.S.). Clinical Pharmacology, The William Harvey Research Institute (O.P., H.R.W., M.J.C.), National Institute for Health Research, Biomedical Research Centre (H.R.W., M.J.C.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom. Department of Pharmacology, University of Oxford, United Kingdom (H.A.L.L., K.A.D., C.J.G.). Medicines Monitoring Unit, School of Medicine (A.D.), Division of Cardiovascular and Diabetes Medicine (C.N.A.), University of Dundee, United Kingdom. Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (J.V.W., J.H., W.F.)
| | - William Fuller
- British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.B., S.T., O.P., O.R., D.S.-T., M.J.S.). Clinical Pharmacology, The William Harvey Research Institute (O.P., H.R.W., M.J.C.), National Institute for Health Research, Biomedical Research Centre (H.R.W., M.J.C.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom. Department of Pharmacology, University of Oxford, United Kingdom (H.A.L.L., K.A.D., C.J.G.). Medicines Monitoring Unit, School of Medicine (A.D.), Division of Cardiovascular and Diabetes Medicine (C.N.A.), University of Dundee, United Kingdom. Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (J.V.W., J.H., W.F.)
| | - Michael J. Shattock
- British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.B., S.T., O.P., O.R., D.S.-T., M.J.S.). Clinical Pharmacology, The William Harvey Research Institute (O.P., H.R.W., M.J.C.), National Institute for Health Research, Biomedical Research Centre (H.R.W., M.J.C.), Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom. Department of Pharmacology, University of Oxford, United Kingdom (H.A.L.L., K.A.D., C.J.G.). Medicines Monitoring Unit, School of Medicine (A.D.), Division of Cardiovascular and Diabetes Medicine (C.N.A.), University of Dundee, United Kingdom. Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (J.V.W., J.H., W.F.)
| |
Collapse
|
3
|
Badimon L, Vilahur G, Rocca B, Patrono C. The key contribution of platelet and vascular arachidonic acid metabolism to the pathophysiology of atherothrombosis. Cardiovasc Res 2021; 117:2001-2015. [PMID: 33484117 DOI: 10.1093/cvr/cvab003] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/17/2020] [Accepted: 01/08/2021] [Indexed: 02/06/2023] Open
Abstract
Arachidonic acid is one of the most abundant and ubiquitous ω-6 polyunsaturated fatty acid, present in esterified form in the membrane phospholipids of all mammalian cells and released from phospholipids by several phospholipases in response to various activating or inhibitory stimuli. Arachidonic acid is the precursor of a large number of enzymatically and non-enzymatically derived, biologically active autacoids, including prostaglandins (PGs), thromboxane (TX) A2, leukotrienes, and epoxyeicosatetraenoic acids (collectively called eicosanoids), endocannabinoids and isoprostanes, respectively. Eicosanoids are local modulators of the physiological functions and pathophysiological roles of blood vessels and platelets. For example, the importance of cyclooxygenase (COX)-1-derived TXA2 from activated platelets in contributing to primary haemostasis and atherothrombosis is demonstrated in animal and human models by the bleeding complications and cardioprotective effects associated with low-dose aspirin, a selective inhibitor of platelet COX-1. The relevance of vascular COX-2-derived prostacyclin (PGI2) in endothelial thromboresistance and atheroprotection is clearly shown by animal and human models and by the adverse cardiovascular effects exerted by COX-2 inhibitors in humans. A vast array of arachidonic acid-transforming enzymes, downstream synthases and isomerases, transmembrane receptors, and specificity in their tissue expression make arachidonic acid metabolism a fine-tuning system of vascular health and disease. Its pharmacological regulation is central in human cardiovascular diseases, as demonstrated by biochemical measurements and intervention trials.
Collapse
Affiliation(s)
- Lina Badimon
- Cardiovascular Program-ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; CIBERCV, Instituto Salud Carlos III, Madrid, Spain.,Cardiovascular Research Chair Autonomous University of Barcelona (UAB), Barcelona, Spain
| | - Gemma Vilahur
- Cardiovascular Program-ICCC, Research Institute-Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; CIBERCV, Instituto Salud Carlos III, Madrid, Spain
| | - Bianca Rocca
- Department of Bioethics and Safety, Section of Pharmacology, Catholic University School of Medicine, Rome, Italy.,Gemelli' Foundation, IRCCS, Rome, Italy
| | - Carlo Patrono
- Department of Bioethics and Safety, Section of Pharmacology, Catholic University School of Medicine, Rome, Italy.,Gemelli' Foundation, IRCCS, Rome, Italy
| |
Collapse
|
4
|
Kamiya T, Omae T, Nakabayashi S, Takahashi K, Tanner A, Yoshida A. Effect of Rho Kinase Inhibitor Ripasudil (K-115) on Isolated Porcine Retinal Arterioles. J Ocul Pharmacol Ther 2020; 37:104-111. [PMID: 33351704 DOI: 10.1089/jop.2020.0082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Purpose: To investigate the vasorelaxation effect of ripasudil (K-115), a novel Rho-associated coiled-coil-containing protein kinase (ROCK) inhibitor, on isolated retinal arterioles. We determined whether the actions of ripasudil on the retinal microvascular diameter were dependent on the endothelium and/or potassium channels in the smooth muscle, with the goals of uncovering the signaling mechanisms required for this vasomotor activity and inhibiting the action of endothelin-1 (ET-1). Methods: In this in vitro study, we isolated porcine retinal arterioles, which were cannulated and pressurized without flow. We recorded diametric changes using videomicroscopic techniques. Results: In a dose-dependent (10 nM-30 μM) manner, retinal arterioles were relaxed in response to ripasudil [maximum % resting diameter, 160.3% ± 7.7% (mean ± standard error of the mean)]. The ripasudil-induced vasorelaxation was unaffected by endothelium removal, using nonselective potassium channel blocker tetraethylammonium, Ca2+-activated large-conductance potassium channel blocker iberiotoxin, voltage-gated potassium channel blocker 4-AP, ATP-sensitive potassium channel blocker glibenclamide, and inward rectifier potassium channel blocker BaCl2. Ripasudil prevented ET-1-caused vasoconstriction of the retinal arterioles regardless of the presence of endothelium to a similar extent. Conclusion: The ROCK inhibitor ripasudil elicits endothelium-independent relaxation and inhibits the action of ET-1 on the retinal arterioles. Determining the relaxation properties of ripasudil on the retinal microvasculature will likely support the development of potential therapies for glaucoma.
Collapse
Affiliation(s)
- Takayuki Kamiya
- Department of Ophthalmology, Asahikawa Medical University, Asahikawa, Japan
| | - Tsuneaki Omae
- Department of Ophthalmology, Asahikawa Medical University, Asahikawa, Japan
| | - Seigo Nakabayashi
- Department of Ophthalmology, Asahikawa Medical University, Asahikawa, Japan
| | - Kengo Takahashi
- Department of Ophthalmology, Asahikawa Medical University, Asahikawa, Japan
| | - Akira Tanner
- Department of Ophthalmology, Asahikawa Medical University, Asahikawa, Japan
| | - Akitoshi Yoshida
- Department of Ophthalmology, Asahikawa Medical University, Asahikawa, Japan
| |
Collapse
|
5
|
Ozen G, Aljesri K, Celik Z, Turkyılmaz G, Turkyılmaz S, Teskin O, Norel X, Topal G. Mechanism of thromboxane receptor-induced vasoconstriction in human saphenous vein. Prostaglandins Other Lipid Mediat 2020; 151:106476. [PMID: 32721526 DOI: 10.1016/j.prostaglandins.2020.106476] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/29/2020] [Accepted: 07/16/2020] [Indexed: 12/18/2022]
Abstract
Saphenous vein (SV) is one of the most widely used graft material in patients undergoing coronary artery bypass graft surgery (CABG). Thromboxane A2 (TXA2) is implicated in graft failure by inducing vasoconstriction and platelet aggregation. The aim of this study is to investigate the mechanism involved in TXA2-induced vasoconstriction in human SV. The role of different inhibitors and blockers on U46619 (TXA2-mimetic)-induced vasoconstriction is investigated by using an isolated organ bath system. Relaxation responses to several mediators are evaluated in SV pre-contracted with U46619 and compared with those pre-contracted with phenylephrine. Our results demonstrate that U46619-induced contraction is completely blocked by myosin light chain kinase inhibitor ML-9 or TP receptor antagonist BAY u3405. Furthermore, U46619-induced contraction is partially inhibited by phospholipase C inhibitor U73122, protein kinase C inhibitor calphostin C, Rho-kinase inhibitor Y-27632, L-type calcium channel blocker nifedipine, store-operated channel inhibitor SKF96365 or removal of extracellular calcium. Relaxation responses to NO donor (sodium nitroprusside), guanylate cyclase (GC) stimulator (riociguat), phosphodiesterase (PDE) inhibitors (sildenafil, IBMX), adenylate cyclase (AC) activator (forskolin) and acetylcholine (ACh) are markedly reduced when U46619 is used as a pre-contraction agent. Our results demonstrate that influx of extracellular Ca2+ (through L-type calcium channels and store-operated calcium channels) and intracellular Ca2+ release together with Ca2+ sensitization (through Rho-kinase activation) are necessary components for TXA2-induced vasoconstriction in SV. Moreover, more pronounced decrease in vasorelaxation induced by several mediators (SNP, riociguat, sildenafil, IBMX, forskolin, and ACh) in the presence of U46619 when compared with phenylephrine suggests that there is a crosstalk between the TP receptor signaling pathway and PDE, AC, GC enzymes. We believe that the investigation of mechanism of the TXA2-induced vasoconstriction in SV will provide additional information for the prevention of SV graft failure.
Collapse
Affiliation(s)
- Gulsev Ozen
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, Istanbul 34116, Turkey.
| | - Khadija Aljesri
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, Istanbul 34116, Turkey
| | - Zeynep Celik
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, Istanbul 34116, Turkey
| | - Gulsum Turkyılmaz
- Department of Cardiovascular Surgery, Bakirkoy Dr Sadi Konuk Education and Research Hospital Bakırkoy, Istanbul, Turkey
| | - Saygın Turkyılmaz
- Department of Cardiovascular Surgery, Bakirkoy Dr Sadi Konuk Education and Research Hospital Bakırkoy, Istanbul, Turkey
| | - Onder Teskin
- Department of Cardiovascular Surgery, Biruni University, Istanbul, Turkey
| | - Xavier Norel
- Université de Paris, INSERM, UMR-S 1148, CHU X. Bichat, 75018 Paris, France
| | - Gokce Topal
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, Istanbul 34116, Turkey
| |
Collapse
|
6
|
Interplay of cardiovascular mediators, oxidative stress and inflammation in liver disease and its complications. Nat Rev Cardiol 2020; 18:117-135. [PMID: 32999450 DOI: 10.1038/s41569-020-0433-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/11/2020] [Indexed: 12/11/2022]
Abstract
The liver is a crucial metabolic organ that has a key role in maintaining immune and endocrine homeostasis. Accumulating evidence suggests that chronic liver disease might promote the development of various cardiac disorders (such as arrhythmias and cardiomyopathy) and circulatory complications (including systemic, splanchnic and pulmonary complications), which can eventually culminate in clinical conditions ranging from portal and pulmonary hypertension to pulmonary, cardiac and renal failure, ascites and encephalopathy. Liver diseases can affect cardiovascular function during the early stages of disease progression. The development of cardiovascular diseases in patients with chronic liver failure is associated with increased morbidity and mortality, and cardiovascular complications can in turn affect liver function and liver disease progression. Furthermore, numerous infectious, inflammatory, metabolic and genetic diseases, as well as alcohol abuse can also influence both hepatic and cardiovascular outcomes. In this Review, we highlight how chronic liver diseases and associated cardiovascular effects can influence different organ pathologies. Furthermore, we explore the potential roles of inflammation, oxidative stress, vasoactive mediator imbalance, dysregulated endocannabinoid and autonomic nervous systems and endothelial dysfunction in mediating the complex interplay between the liver and the systemic vasculature that results in the development of the extrahepatic complications of chronic liver disease. The roles of ageing, sex, the gut microbiome and organ transplantation in this complex interplay are also discussed.
Collapse
|
7
|
Muhammad RN, Sallam N, El-Abhar HS. Activated ROCK/Akt/eNOS and ET-1/ERK pathways in 5-fluorouracil-induced cardiotoxicity: modulation by simvastatin. Sci Rep 2020; 10:14693. [PMID: 32895407 PMCID: PMC7477553 DOI: 10.1038/s41598-020-71531-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 08/18/2020] [Indexed: 12/11/2022] Open
Abstract
5-Fluorouracil (5-FU) is used in the treatment of different solid tumors; however, its use is associated with rare, but serious cardiotoxicity. Nevertheless, the involvement of ROCK/NF-κB, Akt/eNOS and ET-1/ERK1/2 trajectories in the cardiotoxic effect and in the potential cardioprotective upshot of simvastatin has been elusive. Male Wistar rats were allocated into 5-FU (50 mg/kg/week; i.p, 6 weeks), simvastatin (15 mg/kg/day; p.o, 8 weeks) treated groups and simvastatin + 5-FU, besides the normal control group. 5-FU-induced cardiotoxicity boosted the serum level of N-terminal pro-brain (B-type) natriuretic peptide (NT-proBNP), aortic contents of endothelin (ET)-1 and thromboxane (TX) A2, as well as cardiac contents of NADPH oxidases (Nox), cyclooxygenase (COX)-2, malondialdehyde (MDA), phosphorylated Akt (p-Akt), phosphorylated extracellular signal-regulated kinase (p-ERK)1/2 and the protein expressions of rho-kinase (ROCK) and caspase-3. On the other hand, it suppressed cardiac reduced glutathione (GSH) and phosphorylated endothelial nitric oxide synthase (p-eNOS). Contrariwise, co-administration with simvastatin overcame these disturbed events and modulated the ROCK/NF-κB, Akt/eNOS and ET-1/ERK1/2 signaling pathways. This study highlights other mechanisms than coronary artery spasm in the 5-FU cardiotoxicity and reveals that NT-proBNP is a potential early marker in this case. Moreover, the cross-talk between ROCK/ NF-κB, ROS/COX-2/TXA2, Akt/eNOS and ET-1/ERK1/2 pathways contributes via different means to upsetting the vasoconstriction/vasodilatation equilibrium as well as endothelial cell function and finally leads to cardiomyocyte stress and death-the modulation of these trajectories offers simvastatin its potential cardio-protection against 5-FU.
Collapse
Affiliation(s)
- Radwa Nasser Muhammad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Nada Sallam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Hanan Salah El-Abhar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
- Department of Pharmacology & Toxicology, Faculty of Pharmaceutical Sciences and Pharmaceutical Industries, Future University in Egypt, Cairo, 11835, Egypt
| |
Collapse
|
8
|
Shi Y, Leung SWS. Long-term nitric oxide synthase inhibition prevents 17β-estradiol-induced suppression of cyclooxygenase-dependent contractions and enhancement of endothelium-dependent hyperpolarization-like relaxation in mesenteric arteries of ovariectomized rats. Eur J Pharmacol 2020; 882:173275. [PMID: 32535100 DOI: 10.1016/j.ejphar.2020.173275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 10/24/2022]
Abstract
Endothelial dysfunction is associated with a reduced bioavailability of nitric oxide (NO). In this study, the effects of 17β-estradiol supplement on endothelial function were examined in ovariectomized (OVX) rats following long-term inhibition of NO synthases with L-NAME. Female Sprague Dawley rats were ovariectomized at 12 weeks old. They were supplemented with 17β-estradiol (25 μg/kg/day, intramuscularly) or its vehicle (olive oil) until they were killed. At 18 weeks old, they were administered daily with NO synthase inhibitor L-NAME (60 mg/kg, by gavage) or its vehicle (distilled water) for 6 weeks. Rats were then anesthetized for blood pressure measurement and for isolation of mesenteric arteries and aortae for isometric tension measurement. Long-term L-NAME-treatment, without or with 17β-estradiol supplement, resulted in reduced plasma nitrite/nitrate level without causing an increase in blood pressure in OVX rats. Acute inhibition of cyclooxygenase (COX) with indomethacin improved relaxations of mesenteric arteries to the calcium ionophore A23187 in OVX rats, and in those with long-term L-NAME-treatment without or with 17β-estradiol supplement, but not in those with female hormone supplement only. 17β-estradiol supplement or long-term L-NAME-treatment resulted in a greater endothelium-dependent hyperpolarization-like relaxation in mesenteric arteries. In the quiescent aorta, 17β-estradiol supplement or long-term L-NAME-treatment unmasked the COX-dependent components of A23187-induced contractions, but prevented that of the smooth muscle contractions to U46619 in OVX rats. In summary, long-term 17β-estradiol-supplement results in differential effects in different blood vessel types, and its beneficial vascular effects are masked under the conditions with NO synthase inhibition.
Collapse
Affiliation(s)
- Yi Shi
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, PR China
| | - Susan Wai Sum Leung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, PR China.
| |
Collapse
|
9
|
Khan S, Andrews KL, Chin-Dusting JPF. Cyclo-Oxygenase (COX) Inhibitors and Cardiovascular Risk: Are Non-Steroidal Anti-Inflammatory Drugs Really Anti-Inflammatory? Int J Mol Sci 2019; 20:ijms20174262. [PMID: 31480335 PMCID: PMC6747368 DOI: 10.3390/ijms20174262] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 08/08/2019] [Indexed: 12/15/2022] Open
Abstract
Cyclo-oxygenase (COX) inhibitors are among the most commonly used drugs in the western world for their anti-inflammatory and analgesic effects. However, they are also well-known to increase the risk of coronary events. This area is of renewed significance given alarming new evidence suggesting this effect can occur even with acute usage. This contrasts with the well-established usage of aspirin as a mainstay for cardiovascular prophylaxis, as well as overwhelming evidence that COX inhibition induces vasodilation and is protective for vascular function. Here, we present an updated review of the preclinical and clinical literature regarding the cardiotoxicity of COX inhibitors. While studies to date have focussed on the role of COX in influencing renal and vascular function, we suggest an interaction between prostanoids and T cells may be a novel factor, mediating elevated cardiovascular disease risk with NSAID use.
Collapse
Affiliation(s)
- Shanzana Khan
- Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia.
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia.
| | - Karen L Andrews
- Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Jaye P F Chin-Dusting
- Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| |
Collapse
|
10
|
Lagier D, Tonon D, Garrigue P, Guillet B, Giacomino L, Martin JC, Alessi MC, Bruder N, Velly LJ. Thromboxane-prostaglandin receptor antagonist, terutroban, prevents neurovascular events after subarachnoid haemorrhage: a nanoSPECT study in rats. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2019; 23:42. [PMID: 30744667 PMCID: PMC6371436 DOI: 10.1186/s13054-019-2338-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 01/31/2019] [Indexed: 12/12/2022]
Abstract
Background Several lipid metabolites in cerebrospinal fluid are correlated with poor outcomes in aneurysmal subarachnoid haemorrhage. Most of these metabolites bind to ubiquitous thromboxane–prostaglandin (TP) receptors, causing vasoconstriction and inflammation. Here, we evaluated terutroban (TBN), a specific TP receptor antagonist, for the prevention of post-haemorrhage blood-brain barrier disruption, neuronal apoptosis and delayed cerebral hypoperfusion. Methods The rat double subarachnoid haemorrhage model was produced by twice injecting (days 1 and 2) autologous blood into the cisterna magna. Seventy-eight male Sprague-Dawley rats were assigned to experimental groups. Rats exposed to subarachnoid haemorrhage were allocated to no treatment (SAH group) or TBN treatment by gastric gavage during the first 5 days after haemorrhage (SAH+TBN group). Control rats received artificial cerebrospinal fluid injections (CSF group). Sham-operated rats with or without TBN administration were also studied. Body weight and Garcia neurological scores were assessed on day 2 and day 5. We used nanoscale single-photon emission computed tomography (nanoSPECT) to measure brain uptake of three radiolabelled agents: 99mTechnetium-diethylenetriaminepentacetate (99mTc-DTPA), which indicated blood-brain barrier permeability on day 3, 99mTechnetium-annexin V-128 (99mTc-Anx-V128), which indicated apoptosis on day 4, and 99mTechnetium-hexamethylpropyleneamineoxime (99mTc-HMPAO), which indicated cerebral perfusion on day 5. Basilar artery narrowing was verified histologically, and cerebral TP receptor agonists were quantified. Results 99mTc-DTPA uptake unveiled blood-brain barrier disruption in the SAH group. TBN mitigated this disruption in the brainstem area. 99mTc-Anx-V128 uptake was increased in the SAH group and TBN diminished this effect in the cerebellum. 99mTc-HMPAO uptake revealed a global decreased perfusion on day 5 in the SAH group that was significantly counteracted by TBN. TBN also mitigated basilar artery vasoconstriction, neurological deficits (on day 2), body weight loss (on day 5) and cerebral production of vasoconstrictors such as Thromboxane B2 and Prostaglandin F2α. Conclusions Based on in vivo nanoscale imaging, we demonstrated that TBN protected against blood-brain barrier disruption, exerted an anti-apoptotic effect and improved cerebral perfusion. Thus, TP receptor antagonists showed promising results in treating post-haemorrhage neurovascular events.
Collapse
Affiliation(s)
- David Lagier
- Department of Anaesthesiology and Critical Care Medicine, University Hospital Timone, Marseille, France. .,C2VN Inserm 1263, Inra 1260, Aix Marseille University, Marseille, France.
| | - David Tonon
- Department of Anaesthesiology and Critical Care Medicine, University Hospital Timone, Marseille, France.,C2VN Inserm 1263, Inra 1260, Aix Marseille University, Marseille, France
| | - Philippe Garrigue
- CERIMED (European Center for Research in Medical Imaging), Aix Marseille University, Marseille, France
| | - Benjamin Guillet
- CERIMED (European Center for Research in Medical Imaging), Aix Marseille University, Marseille, France
| | - Laura Giacomino
- Department of Anaesthesiology and Critical Care Medicine, INT (Institut de Neurosciences de la Timone), University Hospital Timone, Aix Marseille University, Marseille, France
| | | | | | - Nicolas Bruder
- Department of Anaesthesiology and Critical Care Medicine, University Hospital Timone, Marseille, France
| | - Lionel J Velly
- Department of Anaesthesiology and Critical Care Medicine, INT (Institut de Neurosciences de la Timone), University Hospital Timone, Aix Marseille University, Marseille, France
| |
Collapse
|
11
|
Bassiouni W, Daabees T, Louedec L, Norel X, Senbel A. Evaluation of some prostaglandins modulators on rat corpus cavernosum in-vitro: Is relaxation negatively affected by COX-inhibitors? Biomed Pharmacother 2019; 111:1458-1466. [PMID: 30841461 DOI: 10.1016/j.biopha.2018.12.097] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Prostaglandins (PGs) play an important role in corpus cavernosum relaxation, as evidenced by alprostadil being used as a drug for erectile dysfunction. Reports about the effect of cyclooxygenase (COX) inhibitors on erectile function are highly contradictory. AIM To compare the potential effects of some COX inhibitors with varying COX-1/COX-2 selectivities (indomethacin, ketoprofen and diclofenac) with that of the selective COX-2 inhibitor (DFU) on corpus cavernosal tone in-vitro. The role played by PGE1, PGI2-analogue and PGE4 receptor (EP4)-agonist in controlling corpus cavernosum function and the modulation of their action by sildenafil is also studied. METHODS Organ bath experiments were performed using isolated rat corpus cavernosum. Direct relaxations and changes to electric field stimulation (EFS, 2-16 Hz, 60 V, 0.8 ms, 10 s train)-induced relaxation by the effect of the selected drugs were studied. Strips were precontracted using phenylephrine (PE, 10-5 M). Results are expressed as mean ± SEM of 5-9 rats. RESULTS Alprostadil, iloprost and L902688 (selective EP4 agonist) induced direct relaxation where L902688 showed greater relaxant effect. Sildenafil potentiated the Emax of alprostadil and iloprost but not L902688. EFS and acetylcholine (ACh)-induced relaxations were significantly potentiated in presence of indomethacin, ketoprofen and diclofenac (20, 100 μM) but not in presence of selective COX-2 inhibitor (DFU, 1 μM). GR32191B (Thromboxane A2 receptor antagonist, 10-6 M) significantly reduced the potentiatory effect of indomethacin. Only diclofenac succeeded to potentiate sodium nitroprusside (SNP)-induced relaxation. CONCLUSIONS EP4 receptors may play an important nitric oxide (NO)/cGMP-independent role in corpus cavernosal relaxation. Nonselective COX inhibitors seem of no harm concerning cavernosal tissue relaxation, possibly because they inhibit the synthesis of the highly contracting mediator thromboxane A2.
Collapse
Affiliation(s)
- Wesam Bassiouni
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Egypt
| | - Tahia Daabees
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Egypt
| | - Liliane Louedec
- Laboratory for Vascular Translational Sciences, INSERM U1148, X. Bichat Hospital, University Paris XIII, France
| | - Xavier Norel
- Laboratory for Vascular Translational Sciences, INSERM U1148, X. Bichat Hospital, University Paris XIII, France
| | - Amira Senbel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Egypt; Laboratory for Vascular Translational Sciences, INSERM U1148, X. Bichat Hospital, University Paris XIII, France.
| |
Collapse
|
12
|
Simeone P, Boccatonda A, Liani R, Santilli F. Significance of urinary 11-dehydro-thromboxane B 2 in age-related diseases: Focus on atherothrombosis. Ageing Res Rev 2018; 48:51-78. [PMID: 30273676 DOI: 10.1016/j.arr.2018.09.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/13/2018] [Accepted: 09/23/2018] [Indexed: 12/13/2022]
Abstract
Platelet activation plays a key role in atherogenesis and atherothrombosis. Biochemical evidence of increased platelet activation in vivo can be reliably obtained through non-invasive measurement of thromboxane metabolite (TXM) excretion. Persistent biosynthesis of TXA2 has been associated with several ageing-related diseases, including acute and chronic cardio-cerebrovascular diseases and cardiovascular risk factors, such as cigarette smoking, type 1 and type 2 diabetes mellitus, obesity, hypercholesterolemia, hyperhomocysteinemia, hypertension, chronic kidney disease, chronic inflammatory diseases. Given the systemic nature of TX excretion, involving predominantly platelet but also extraplatelet sources, urinary TXM may reflect either platelet cyclooxygenase-1 (COX-1)-dependent TX generation or COX-2-dependent biosynthesis by inflammatory cells and/or platelets, or a combination of the two, especially in clinical settings characterized by low-grade inflammation or enhanced platelet turnover. Although urinary 11-dehydro-TXB2 levels are largely suppressed with low-dose aspirin, incomplete TXM suppression by aspirin predicts the future risk of vascular events and death in high-risk patients and may identify individuals who might benefit from treatments that more effectively block in vivo TX production or activity. Several disease-modifying agents, including lifestyle intervention, antidiabetic drugs and antiplatelet agents besides aspirin have been shown to reduce TX biosynthesis. Taken together, these aspects may contribute to the development of promising mechanism-based therapeutic strategies to reduce the progression of atherothrombosis. We intended to critically review current knowledge on both the pathophysiological significance of urinary TXM excretion in clinical settings related to ageing and atherothrombosis, as well as its prognostic value as a biomarker of vascular events.
Collapse
Affiliation(s)
- Paola Simeone
- Department of Medicine and Aging, and Center of Aging Science and Translational Medicine (CESI-Met), Via Luigi Polacchi, Chieti, Italy
| | - Andrea Boccatonda
- Department of Medicine and Aging, and Center of Aging Science and Translational Medicine (CESI-Met), Via Luigi Polacchi, Chieti, Italy
| | - Rossella Liani
- Department of Medicine and Aging, and Center of Aging Science and Translational Medicine (CESI-Met), Via Luigi Polacchi, Chieti, Italy
| | - Francesca Santilli
- Department of Medicine and Aging, and Center of Aging Science and Translational Medicine (CESI-Met), Via Luigi Polacchi, Chieti, Italy.
| |
Collapse
|
13
|
Pál É, Hadjadj L, Fontányi Z, Monori-Kiss A, Mezei Z, Lippai N, Magyar A, Heinzlmann A, Karvaly G, Monos E, Nádasy G, Benyó Z, Várbíró S. Vitamin D deficiency causes inward hypertrophic remodeling and alters vascular reactivity of rat cerebral arterioles. PLoS One 2018; 13:e0192480. [PMID: 29408903 PMCID: PMC5800593 DOI: 10.1371/journal.pone.0192480] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 01/24/2018] [Indexed: 02/07/2023] Open
Abstract
Background and purpose Vitamin D deficiency (VDD) is a global health problem, which can lead to several pathophysiological consequences including cardiovascular diseases. Its impact on the cerebrovascular system is not well understood. The goal of the present work was to examine the effects of VDD on the morphological, biomechanical and functional properties of cerebral arterioles. Methods Four-week-old male Wistar rats (n = 11 per group) were either fed with vitamin D deficient diet or received conventional rat chow with per os vitamin D supplementation. Cardiovascular parameters and hormone levels (testosterone, androstenedione, progesterone and 25-hydroxyvitamin D) were measured during the study. After 8 weeks of treatment anterior cerebral artery segments were prepared and their morphological, biomechanical and functional properties were examined using pressure microangiometry. Resorcin-fuchsin and smooth muscle actin staining were used to detect elastic fiber density and smooth muscle cell counts in the vessel wall, respectively. Sections were immunostained for eNOS and COX-2 as well. Results VDD markedly increased the wall thickness, the wall-to-lumen ratio and the wall cross-sectional area of arterioles as well as the number of smooth muscle cells in the tunica media. As a consequence, tangential wall stress was significantly lower in the VDD group. In addition, VDD increased the myogenic as well as the uridine 5’-triphosphate-induced tone and impaired bradykinin-induced relaxation. Decreased eNOS and increased COX-2 expression were also observed in the endothelium of VDD animals. Conclusions VDD causes inward hypertrophic remodeling due to vascular smooth muscle cell proliferation and enhances the vessel tone probably because of increased vasoconstrictor prostanoid levels in young adult rats. In addition, the decreased eNOS expression results in endothelial dysfunction. These morphological and functional alterations can potentially compromise the cerebral circulation and lead to cerebrovascular disorders in VDD.
Collapse
Affiliation(s)
- Éva Pál
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
- * E-mail:
| | - Leila Hadjadj
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Zoltán Fontányi
- 2 Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| | - Anna Monori-Kiss
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Zsuzsanna Mezei
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Norbert Lippai
- Department of Pathology, Jász-Nagykun-Szolnok County Hetényi Géza Hospital, Szolnok, Hungary
| | - Attila Magyar
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Andrea Heinzlmann
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Gellért Karvaly
- Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary
- Bionics Innovation Center, Budapest, Hungary
| | - Emil Monos
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - György Nádasy
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Szabolcs Várbíró
- 2 Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
14
|
Silva B, Pernomian L, De Paula T, Grando M, Bendhack L. Endothelial nitric oxide synthase and cyclooxygenase are activated by hydrogen peroxide in renal hypertensive rat aorta. Eur J Pharmacol 2017; 814:87-94. [DOI: 10.1016/j.ejphar.2017.07.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/28/2017] [Accepted: 07/28/2017] [Indexed: 10/19/2022]
|
15
|
Abstract
PURPOSE OF REVIEW Endothelial dysfunction is intimately related to the development of various cardiovascular diseases, including hypertension, and is often used as a target for pharmacological treatment. The scope of this review is to assess effects of aspirin on endothelial function and their clinical implication in arterial hypertension. RECENT FINDINGS Emerging data indicate the role of platelets in the development of vascular inflammation due to the release of proinflammatory mediators, for example, triggered largely by thromboxane. Vascular inflammation further promotes oxidative stress, diminished synthesis of vasodilators, proaggregatory and procoagulant state. These changes translate into vasoconstriction, impaired circulation and thrombotic complications. Aspirin inhibits thromboxane synthesis, abolishes platelets activation and acetylates enzymes switching them to the synthesis of anti-inflammatory substances. Aspirin pleiotropic effects have not been fully elucidated yet. In secondary prevention studies, the decrease in cardiovascular events with aspirin outweighs bleeding risks, but this is not the case in primary prevention settings. Ongoing trials will provide more evidence on whether to expand the use of aspirin or stay within current recommendations.
Collapse
Affiliation(s)
- Mikhail S Dzeshka
- University of Birmingham Institute of Cardiovascular Sciences, City Hospital, Dudley Road, Birmingham, B18 7QH, UK
- Grodno State Medical University, Grodno, Belarus
| | - Alena Shantsila
- University of Birmingham Institute of Cardiovascular Sciences, City Hospital, Dudley Road, Birmingham, B18 7QH, UK
| | - Gregory Y H Lip
- University of Birmingham Institute of Cardiovascular Sciences, City Hospital, Dudley Road, Birmingham, B18 7QH, UK.
- Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
16
|
Romero M, Leon-Gomez E, Lobysheva I, Rath G, Dogné JM, Feron O, Dessy C. Effects of BM-573 on Endothelial Dependent Relaxation and Increased Blood Pressure at Early Stages of Atherosclerosis. PLoS One 2016; 11:e0152579. [PMID: 27019366 PMCID: PMC4809599 DOI: 10.1371/journal.pone.0152579] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 03/16/2016] [Indexed: 12/19/2022] Open
Abstract
Endothelial dysfunction is considered to be an early event in atherosclerosis and plays a pivotal role in the development, progression and clinical complications of atherosclerosis. Previous studies have shown the beneficial effects of combined inhibition of thromboxane synthase and antagonism of thromboxane receptors by BM-573 on atherosclerosis; however our knowledge about the beneficial effects of BM-573 on endothelial function and increased blood pressure related to early stage of atherosclerosis is limited. In the present study, we investigated the effects of short-term (3 μM, 1 hour) and chronic (10 mg/L, 8 weeks) treatments with BM-573 on vasodilatory function, nitric oxide (NO) bioavailability, oxidative stress and systolic blood pressure in 15 weeks old apolipoprotein E-deficient (ApoE-KO) mice. ApoE-KO mice showed a reduced endothelium-derived relaxation. In addition, NO bioavailability was reduced and oxidative stress and blood pressure were increased in ApoE-KO mice versus wild-type mice. BM-573 treatments were able to improve the relaxation profile in ApoE-KO mice. Short-term effects of BM-573 were mainly mediated by an increased phosphorylation of both eNOS and Akt, whereas BM-573 in vivo treatment also reduced oxidative stress and restored NO bioavailability. In addition, chronic administration of BM-573 reduced systolic blood pressure in ApoE-KO mice. In conclusion, pharmacological modulation of TxA2 biosynthesis and biological activities by dual TP antagonism/TxAS inhibition with BM-573, already known to prevent plaque formation, has the potential to correct vasodilatory dysfunction at the early stages of atherosclerosis.
Collapse
Affiliation(s)
- Miguel Romero
- Pole of Pharmacology and Therapeutics (FATH), Institute of Experimental & Clinical Research (IREC), Université Catholique de Louvain (UCL) Medical School, Brussels, Belgium
- * E-mail: (MR); (CD)
| | - Elvira Leon-Gomez
- Pole of Pharmacology and Therapeutics (FATH), Institute of Experimental & Clinical Research (IREC), Université Catholique de Louvain (UCL) Medical School, Brussels, Belgium
| | - Irina Lobysheva
- Pole of Pharmacology and Therapeutics (FATH), Institute of Experimental & Clinical Research (IREC), Université Catholique de Louvain (UCL) Medical School, Brussels, Belgium
| | - Géraldine Rath
- Pole of Pharmacology and Therapeutics (FATH), Institute of Experimental & Clinical Research (IREC), Université Catholique de Louvain (UCL) Medical School, Brussels, Belgium
| | | | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institute of Experimental & Clinical Research (IREC), Université Catholique de Louvain (UCL) Medical School, Brussels, Belgium
| | - Chantal Dessy
- Pole of Pharmacology and Therapeutics (FATH), Institute of Experimental & Clinical Research (IREC), Université Catholique de Louvain (UCL) Medical School, Brussels, Belgium
- * E-mail: (MR); (CD)
| |
Collapse
|
17
|
Vidal-Gómez X, Novella S, Pérez-Monzó I, Garabito M, Dantas AP, Segarra G, Hermenegildo C, Medina P. Decreased bioavailability of nitric oxide in aorta from ovariectomized senescent mice. Role of cyclooxygenase. Exp Gerontol 2016; 76:1-8. [PMID: 26774228 DOI: 10.1016/j.exger.2016.01.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 12/23/2015] [Accepted: 01/12/2016] [Indexed: 01/22/2023]
Abstract
This study investigates the effects of aging and/or ovariectomy on vascular reactivity to thromboxane A2 (TXA2) receptor stimulation with U46619, and the modulation by nitric oxide (NO) and cyclooxygenase (COX) in aorta from female senescence-accelerated mice (SAMP8) and from senescence resistant mice (SAMR1). Five-month-old female SAMR1 and SAMP8 were divided into three groups: sham-operated, ovariectomized and ovariectomized plus estradiol. Twenty-eight days after surgery, thoracic aortic rings were mounted for isometric recording of tension and concentration-response curves for U46619 (10(-10)-3 × 10(-7) M) were performed in the absence and in the presence of the NO synthase inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME, 10(-4) M) and/or COX inhibitor indomethacin (10(-5)M). Vascular superoxide production was detected by dihydroethidium staining on sections of thoracic aorta. NO bioavailability in response to U46619 was suppressed by estrogen withdrawn in young and senescent mice and was restored by the administration of estradiol. In the presence of indomethacin, contractions to U46619 decreased in all groups indicating an aging- and estrogen-dependent modulation of contractile prostanoids. The simultaneous incubation of L-NAME and indomethacin did not change the maximal responses and sensitivities to TXA2 in any group in comparison with untreated aortic segments. The superoxide generation induced by TXA2 was greater in aorta from SAMP8 than in SAMR1. Moreover, in ovariectomized groups superoxide production was further increased and treatment with 17β-estradiol reverted the effects of the ovariectomy. Inhibition of COX with indomethacin prevented the U46619-induced increase in superoxide formation. Our results indicate that NO bioavailability in response to TP receptor activation is both estrogen- and aging-dependent. TXA2 induced contractions are partially mediated by COX activation. Both aging and ovariectomy enhanced COX-dependent component of the TXA2-induced contraction. It is noteworthy that in the absence of estrogen, COX inhibition induces an increase of NO bioavailability. Therefore, in senescent female mice with an experimental menopause, TP-receptor stimulation is responsible for COX activation and enhanced superoxide generation, which may result in reduced NO bioavailability. These effects were reversed by estrogen administration.
Collapse
Affiliation(s)
- Xavier Vidal-Gómez
- Department of Physiology, University of Valencia, Valencia, Spain; INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Susana Novella
- Department of Physiology, University of Valencia, Valencia, Spain; INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Isabel Pérez-Monzó
- Department of Physiology, University of Valencia, Valencia, Spain; INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Manel Garabito
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Institut Clinic del Tòrax, Barcelona, Spain
| | - Ana Paula Dantas
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Institut Clinic del Tòrax, Barcelona, Spain
| | - Gloria Segarra
- Department of Physiology, University of Valencia, Valencia, Spain; INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Carlos Hermenegildo
- Department of Physiology, University of Valencia, Valencia, Spain; INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Pascual Medina
- Department of Physiology, University of Valencia, Valencia, Spain; INCLIVA Biomedical Research Institute, Valencia, Spain.
| |
Collapse
|
18
|
Matsumoto T, Goulopoulou S, Taguchi K, Tostes RC, Kobayashi T. Constrictor prostanoids and uridine adenosine tetraphosphate: vascular mediators and therapeutic targets in hypertension and diabetes. Br J Pharmacol 2015; 172:3980-4001. [PMID: 26031319 DOI: 10.1111/bph.13205] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 04/16/2015] [Accepted: 05/19/2015] [Indexed: 12/22/2022] Open
Abstract
Vascular dysfunction plays a pivotal role in the development of systemic complications associated with arterial hypertension and diabetes. The endothelium, or more specifically, various factors derived from endothelial cells tightly regulate vascular function, including vascular tone. In physiological conditions, there is a balance between endothelium-derived factors, that is, relaxing factors (endothelium-derived relaxing factors; EDRFs) and contracting factors (endothelium-derived contracting factors; EDCFs), which mediate vascular homeostasis. However, in disease states, such as diabetes and arterial hypertension, there is an imbalance between EDRF and EDCF, with a reduction of EDRF signalling and an increase of EDCF signalling. Among EDCFs, COX-derived vasoconstrictor prostanoids play an important role in the development of vascular dysfunction associated with hypertension and diabetes. Moreover, uridine adenosine tetraphosphate (Up4 A), identified as an EDCF in 2005, also modulates vascular function. However, the role of Up4 A in hypertension- and diabetes-associated vascular dysfunction is unclear. In the present review, we focused on experimental and clinical evidence that implicate these two EDCFs (vasoconstrictor prostanoids and Up4 A) in vascular dysfunction associated with hypertension and diabetes.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Styliani Goulopoulou
- Department of Integrative Physiology and Anatomy, Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, Japan
| |
Collapse
|
19
|
Vairappan B. Endothelial dysfunction in cirrhosis: Role of inflammation and oxidative stress. World J Hepatol 2015; 7:443-459. [PMID: 25848469 PMCID: PMC4381168 DOI: 10.4254/wjh.v7.i3.443] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 11/08/2014] [Accepted: 12/01/2014] [Indexed: 02/06/2023] Open
Abstract
This review describes the recent developments in the pathobiology of endothelial dysfunction (ED) in the context of cirrhosis with portal hypertension and defines novel strategies and potential targets for therapy. ED has prognostic implications by predicting unfavourable early hepatic events and mortality in patients with portal hypertension and advanced liver diseases. ED characterised by an impaired bioactivity of nitric oxide (NO) within the hepatic circulation and is mainly due to decreased bioavailability of NO and accelerated degradation of NO with reactive oxygen species. Furthermore, elevated inflammatory markers also inhibit NO synthesis and causes ED in cirrhotic liver. Therefore, improvement of NO availability in the hepatic circulation can be beneficial for the improvement of endothelial dysfunction and associated portal hypertension in patients with cirrhosis. Furthermore, therapeutic agents that are identified in increasing NO bioavailability through improvement of hepatic endothelial nitric oxide synthase (eNOS) activity and reduction in hepatic asymmetric dimethylarginine, an endogenous modulator of eNOS and a key mediator of elevated intrahepatic vascular tone in cirrhosis would be interesting therapeutic approaches in patients with endothelial dysfunction and portal hypertension in advanced liver diseases.
Collapse
|
20
|
Stanley C, O'Sullivan SE. Vascular targets for cannabinoids: animal and human studies. Br J Pharmacol 2014; 171:1361-78. [PMID: 24329566 DOI: 10.1111/bph.12560] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 10/18/2013] [Accepted: 11/18/2013] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Application of cannabinoids and endocannabinoids to perfused vascular beds or individual isolated arteries results in changes in vascular resistance. In most cases, the result is vasorelaxation, although vasoconstrictor responses are also observed. Cannabinoids also modulate the actions of vasoactive compounds including acetylcholine, methoxamine, angiotensin II and U46619 (thromboxane mimetic). Numerous mechanisms of action have been proposed including receptor activation, potassium channel activation, calcium channel inhibition and the production of vasoactive mediators such as calcitonin gene-related peptide, prostanoids, NO, endothelial-derived hyperpolarizing factor and hydrogen peroxide. The purpose of this review is to examine the evidence for the range of receptors now known to be activated by cannabinoids. Direct activation by cannabinoids of CB1 , CBe , TRPV1 (and potentially other TRP channels) and PPARs in the vasculature has been observed. A potential role for CB2, GPR55 and 5-HT1 A has also been identified in some studies. Indirectly, activation of prostanoid receptors (TP, IP, EP1 and EP4 ) and the CGRP receptor is involved in the vascular responses to cannabinoids. The majority of this evidence has been obtained through animal research, but recent work has confirmed some of these targets in human arteries. Vascular responses to cannabinoids are enhanced in hypertension and cirrhosis, but are reduced in obesity and diabetes, both due to changes in the target sites of action. Much further work is required to establish the extent of vascular actions of cannabinoids and the application of this research in physiological and pathophysiological situations. LINKED ARTICLES This article is part of a themed section on Cannabinoids 2013. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-6.
Collapse
Affiliation(s)
- Christopher Stanley
- School of Graduate Entry Medicine and Health, University of Nottingham, Royal Derby Hospital, Derby, UK
| | | |
Collapse
|
21
|
Liu C, Bai Y, Xu X, Sun L, Wang A, Wang TY, Maurya SK, Periasamy M, Morishita M, Harkema J, Ying Z, Sun Q, Rajagopalan S. Exaggerated effects of particulate matter air pollution in genetic type II diabetes mellitus. Part Fibre Toxicol 2014; 11:27. [PMID: 24886175 PMCID: PMC4049808 DOI: 10.1186/1743-8977-11-27] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 05/16/2014] [Indexed: 12/14/2022] Open
Abstract
Background Prior experimental and epidemiologic data support a link between exposure to fine ambient particulate matter (<2.5 μm in aerodynamic diameter, PM2.5) and development of insulin resistance/Type II diabetes mellitus. This study was designed to investigate whether inhalational exposure of concentrated PM2.5 in a genetically susceptible animal model would result in abnormalities in energy metabolism and exacerbation of peripheral glycemic control. Methods KKay mice, which are susceptible to Type II DM, were assigned to either concentrated ambient PM2.5 or filtered air (FA) for 5–8 weeks via a whole body exposure system. Glucose tolerance, insulin sensitivity, oxygen consumption and heat production were evaluated. At euthanasia, blood, spleen and visceral adipose tissue were collected to measure inflammatory cells using flow cytometry. Standard immnunohistochemical methods, western blotting and quantitative PCR were used to assess targets of interest. Results PM2.5 exposure influenced energy metabolism including O2 consumption, CO2 production, respiratory exchange ratio and thermogenesis. These changes were accompanied by worsened insulin resistance, visceral adiposity and inflammation in spleen and visceral adipose depots. Plasma adiponectin were decreased in response to PM2.5 exposure while leptin levels increased. PM2.5 exposure resulted in a significant increase in expression of inflammatory genes and decreased UCP1 expression in brown adipose tissue and activated p38 and ERK pathways in the liver of the KKay mice. Conclusions Concentrated ambient PM2.5 exposure impairs energy metabolism, concomitant with abnormalities in glucose homeostasis, increased inflammation in insulin responsive organs, brown adipose inflammation and results in imbalance in circulating leptin/adiponectin levels in a genetically susceptible diabetic model. These results provide additional insights into the mechanisms surrounding air pollution mediated susceptibility to Type II DM.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Sanjay Rajagopalan
- Division of Cardiovascular Medicine, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
22
|
Liu C, Xu X, Bai Y, Wang TY, Rao X, Wang A, Sun L, Ying Z, Gushchina L, Maiseyeu A, Morishita M, Sun Q, Harkema JR, Rajagopalan S. Air pollution-mediated susceptibility to inflammation and insulin resistance: influence of CCR2 pathways in mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2014; 122:17-26. [PMID: 24149114 PMCID: PMC3888572 DOI: 10.1289/ehp.1306841] [Citation(s) in RCA: 153] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 09/27/2013] [Indexed: 05/19/2023]
Abstract
BACKGROUND Epidemiologic and experimental studies support an association between PM2.5 exposure and insulin resistance (IR). Innate immune cell activation has been suggested to play a role in the pathogenesis of these effects. OBJECTIVES We sought to evaluate the role of CC-chemokine receptor 2 (CCR2) in PM2.5-mediated inflammation and IR. METHODS Wild-type C57BL/6 and CCR2-/- male mice were fed a high-fat diet and exposed to either concentrated ambient PM2.5 or filtered air for 17 weeks via a whole-body exposure system. We evaluated glucose tolerance and insulin sensitivity. At euthanasia, blood, spleen, and visceral adipose tissue (VAT) were collected, and inflammatory cells were measured using flow cytometry. We used standard immunoblots, immunohistochemical methods, and quantitative PCR (polymerase chain reaction) to assess pathways of interest involving insulin signaling, inflammation, and lipid and glucose metabolism in various organs. Vascular function was assessed using myography. RESULTS PM2.5 exposure resulted in whole-body IR and increased hepatic lipid accumulation in the liver, which was attenuated in CCR2-/- mice by inhibiting SREBP1c-mediated transcriptional programming, decreasing fatty acid uptake, and suppressing p38 MAPK activity. Abnormal phosphorylation levels of AKT, AMPK in VAT, and adipose tissue macrophage content in wild-type mice were not present in CCR2-/- mice. However, the impaired whole-body glucose tolerance and reduced GLUT-4 in skeletal muscle in response to PM2.5 was not corrected by CCR2 deficiency. CONCLUSIONS PM2.5 mediates IR by regulating VAT inflammation, hepatic lipid metabolism, and glucose utilization in skeletal muscle via both CCR2-dependent and -independent pathways. These findings provide new mechanistic links between air pollution and metabolic abnormalities underlying IR.
Collapse
Affiliation(s)
- Cuiqing Liu
- Department of Physiology, Hangzhou Normal University, Hangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Targeting of Rho kinase ameliorates impairment of diabetic endothelial function in intrarenal artery. Int J Mol Sci 2013; 14:20282-98. [PMID: 24129169 PMCID: PMC3821615 DOI: 10.3390/ijms141020282] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 07/11/2013] [Accepted: 09/09/2013] [Indexed: 02/04/2023] Open
Abstract
Endothelial dysfunction in kidney vasculature is the initial and key element for nephropathy in diabetes mellitus. Accumulating evidence suggests the protective role of Rho kinase inhibitors in endothelial dysfunction via modulating eNOS activity and NO production. However, the role of Rho kinase in diabetes-related endothelial dysfunction in kidney vasculature and the relevant mechanisms remain unknown. We assessed whether pharmacological inhibition of Rho kinase attenuates endothelial dysfunction in intrarenal arteries from type 1 diabetic rats. Fasudil, a Rho kinase inhibitor effectively decreased the phosphorylated level of MYPT1 without affecting the expression of ROCKs in the kidney. Fasudil treatment showed no improvement in diabetes-related abnormality in metabolic indices, but it significantly ameliorated endothelial dysfunction in intrarenal arteries and lessened the mesangial matrix expansion in the kidney cortex. Mechanistically, superoxide production in the intrarenal artery and NOX4 member of NADPH oxidase in the renal cortex that contribute to diabetic nephropathy were also prevented by the Rho kinase inhibitor. In conclusion, the present results indicate that Rho kinase is involved in endothelial dysfunction in type 1 diabetes via enhancement of oxidative stress and provides new evidence for Rho kinase inhibitors as potential therapeutic agents for the treatment of diabetic nephropathy.
Collapse
|
24
|
Rosado E, Rodríguez-Vilarrupla A, Gracia-Sancho J, Tripathi D, García-Calderó H, Bosch J, García-Pagán JC. Terutroban, a TP-receptor antagonist, reduces portal pressure in cirrhotic rats. Hepatology 2013; 58:1424-35. [PMID: 23703868 DOI: 10.1002/hep.26520] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 04/12/2013] [Accepted: 05/07/2013] [Indexed: 12/11/2022]
Abstract
UNLABELLED Increased production of vasoconstrictive prostanoids, such as thromboxane A2 (TXA2 ), contributes to endothelial dysfunction and increased hepatic vascular tone in cirrhosis. TXA2 induces vasoconstriction by way of activation of the thromboxane-A2 /prostaglandin-endoperoxide (TP) receptor. This study investigated whether terutroban, a specific TP receptor blocker, decreases hepatic vascular tone and portal pressure in rats with cirrhosis due to carbon tetrachloride (CCl4 ) or bile duct ligation (BDL). Hepatic and systemic hemodynamics, endothelial dysfunction, liver fibrosis, hepatic Rho-kinase activity (a marker of hepatic stellate cell contraction), and the endothelial nitric oxide synthase (eNOS) signaling pathway were measured in CCl4 and BDL cirrhotic rats treated with terutroban (30 mg/kg/day) or its vehicle for 2 weeks. Terutroban reduced portal pressure in both models without producing significant changes in portal blood flow, suggesting a reduction in hepatic vascular resistance. Terutroban did not significantly change arterial pressure in CCl4 -cirrhotic rats but decreased it significantly in BDL-cirrhotic rats. In livers from CCl4 and BDL-cirrhotic terutroban-treated rats, endothelial dysfunction was improved and Rho-kinase activity was significantly reduced. In CCl4 -cirrhotic rats, terutroban reduced liver fibrosis and decreased alpha smooth muscle actin (α-SMA), collagen-I, and transforming growth factor beta messenger RNA (mRNA) expression without significant changes in the eNOS pathway. In contrast, no change in liver fibrosis was observed in BDL-cirrhotic rats but an increase in the eNOS pathway. CONCLUSION Our data indicate that TP-receptor blockade with terutroban decreases portal pressure in cirrhosis. This effect is due to decreased hepatic resistance, which in CCl4 -cirrhotic rats was linked to decreased hepatic fibrosis, but not in BDL rats, in which the main mediator appeared to be an enhanced eNOS-dependent vasodilatation, which was not liver-selective, as it was associated with decreased arterial pressure. The potential use of terutroban for portal hypertension requires further investigation.
Collapse
Affiliation(s)
- Eugenio Rosado
- Hepatic Hemodynamic Laboratory, Liver Unit, IMDIM, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Ciberehd, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
25
|
Rosado E, Rodríguez-Vilarrupla A, Gracia-Sancho J, Monclús M, Bosch J, García-Pagán JC. Interaction between NO and COX pathways modulating hepatic endothelial cells from control and cirrhotic rats. J Cell Mol Med 2013; 16:2461-70. [PMID: 22436078 PMCID: PMC3823440 DOI: 10.1111/j.1582-4934.2012.01563.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Reduced intrahepatic nitric oxide (NO) bioavailability and increased cyclooxygenase-1 (COX-1)-derived vasoconstrictor prostanoids modulate the hepatic vascular tone in cirrhosis. We aimed at investigating the reciprocal interactions between NO and COX in the hepatic endothelium of control and cirrhotic rats. NO bioavailability (DAF-FM-DA staining), superoxide (O2−) content (DHE staining), prostanoid production (PGI2 and TXA2 by enzyme immunoassays) as well as COX expression (Western Blot), were determined in hepatic endothelial cells (HEC) from control and cirrhotic rats submitted to different experimental conditions: COX activation, COX inhibition, NO activation and NO inhibition. In control and cirrhotic HEC, COX activation with arachidonic acid reduced NO bioavailability and increased O2− levels. These effects were abolished by pre-treating HEC with the COX inhibitor indomethacin. In control, but not in cirrhotic HEC, scavenging of O2− by superoxide dismutase (SOD) incubation partially restored the decrease in NO bioavailability promoted by COX activation. NO supplementation produced a significant and parallel reduction in PGI2 and TXA2 production in control HEC, whereas it only reduced TXA2 production in cirrhotic HEC. By contrast, in control and cirrhotic HEC, NO inhibition did not modify COX expression or activity. Our results demonstrate that NO and COX systems are closely interrelated in HEC. This is especially relevant in cirrhotic HEC where COX inhibition increases NO bioavailability and NO supplementation induces a reduction in TXA2. These strategies may have beneficial effects ameliorating the vasoconstrictor/vasodilator imbalance of the intrahepatic circulation of cirrhotic livers.
Collapse
Affiliation(s)
- Eugenio Rosado
- Hepatic Hemodynamic Laboratory, Liver Unit, IMDIM, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Ciberehd, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
26
|
Dong J, Wong SL, Lau CW, Liu J, Wang YX, Dan He Z, Fai Ng C, Yu Chen Z, Yao X, Xu A, Ni X, Wang H, Huang Y. Calcitriol restores renovascular function in estrogen-deficient rats through downregulation of cyclooxygenase-2 and the thromboxane-prostanoid receptor. Kidney Int 2013; 84:54-63. [PMID: 23423254 DOI: 10.1038/ki.2013.12] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Revised: 11/19/2012] [Accepted: 11/30/2012] [Indexed: 01/18/2023]
Abstract
Cardiovascular risks increase in postmenopausal women. While vitamin D is supplemented for osteoporosis, it is not known whether it protects renal arterial function during estrogen deficiency. Here we measured changes in renovascular reactivity induced by ovariectomy in rats and examined whether calcitriol, the most active form of vitamin D, was able to correct such changes. The impairment of endothelium-dependent relaxation in renal arteries from ovariectomized rats was effectively reversed by long-term calcitriol treatment. It was also corrected by acute exposure to cyclooxygenase-2 (COX-2) inhibitors and a thromboxane-prostanoid receptor antagonist, respectively. Calcitriol normalized the overexpression of COX-2 and thromboxane-prostanoid receptors in intralobal renal artery segments and aortic endothelial cells isolated from ovariectomized rats. In vitro exposure of the arterial segments to calcitriol for 12 h improved relaxation and downregulated thromboxane-prostanoid receptors. The attenuated nitric oxide production in ovariectomized rat aortic endothelial cells was restored following a 12-h treatment with calcitriol, COX-2 inhibition, or thromboxane-prostanoid receptor antagonism. Thus, impaired endothelium-dependent renal artery relaxation in ovariectomized rats is mediated largely through increased activity and expression of COX-2 and the thromboxane-prostanoid receptor. Calcitriol restores endothelial function through downregulating both signaling proteins during estrogen deficiency.
Collapse
Affiliation(s)
- Jinghui Dong
- Institute of Vascular Medicine, Li Ka Shing Institute of Health Sciences, and School of Biomedical Sciences, Hong Kong, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Liu C, Desikan R, Ying Z, Gushchina L, Kampfrath T, Deiuliis J, Wang A, Xu X, Zhong J, Rao X, Sun Q, Maiseyeu A, Parthasarathy S, Rajagopalan S. Effects of a novel pharmacologic inhibitor of myeloperoxidase in a mouse atherosclerosis model. PLoS One 2012; 7:e50767. [PMID: 23251382 PMCID: PMC3519467 DOI: 10.1371/journal.pone.0050767] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 10/24/2012] [Indexed: 11/24/2022] Open
Abstract
Inflammation and oxidative stress play fundamental roles in the pathogenesis of atherosclerosis. Myeloperoxidase has been extensively implicated as a key mediator of inflammatory and redox-dependent processes in atherosclerosis. However, the effect of synthetic myeloperoxidase inhibitors on atherosclerosis has been insufficiently studied. In this study, ApoE(-/-) mice were randomized to low- and high-dose INV-315 groups for 16 weeks on high-fat diet. INV-315 resulted in reduced plaque burden and improved endothelial function in response to acetylcholine. These effects occurred without adverse events or changes in body weight or blood pressure. INV-315 treatment resulted in a decrease in iNOS gene expression, superoxide production and nitrotyrosine content in the aorta. Circulating IL-6 and inflammatory CD11b(+)/Ly6G(low)/7/4(hi) monocytes were significantly decreased in response to INV-315 treatment. Acute pretreatment with INV-315 blocked TNFα-mediated leukocyte adhesion in cremasteric venules and inhibited myeloperoxidase activity. Cholesterol efflux was significantly increased by high-dose INV-315 via ex-vivo reverse cholesterol transport assays. Our results suggest that myeloperoxidase inhibition may exert anti-atherosclerotic effects via inhibition of oxidative stress and enhancement of cholesterol efflux. These findings demonstrate a role for pharmacologic modulation of myeloperoxidase in atherosclerosis.
Collapse
Affiliation(s)
- Cuiqing Liu
- Department of Physiology, Hangzhou Normal University, Hangzhou, China
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | | | - Zhekang Ying
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Liubov Gushchina
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Thomas Kampfrath
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Jeffrey Deiuliis
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Aixia Wang
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Xiaohua Xu
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Jixin Zhong
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Xiaoquan Rao
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Qinghua Sun
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Andrei Maiseyeu
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Sampath Parthasarathy
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Sanjay Rajagopalan
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
28
|
Grad E, Pachino RM, FitzGerald GA, Danenberg HD. Role of Thromboxane Receptor in C-Reactive Protein–Induced Thrombosis. Arterioscler Thromb Vasc Biol 2012; 32:2468-74. [DOI: 10.1161/atvbaha.112.256073] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Objective—
Thromboxane A
2
and prostacyclin are thromboregulatory prostaglandins. The inflammatory C-reactive protein (CRP) promotes thrombosis after vascular injury, presumably via potentiation of thromboxane activity. Using a genetic approach, we investigated the role of thromboxane receptor (TP) pathway in CRP-induced thrombosis.
Methods and Results—
Four genetically engineered mice strains were used:
C57BL
/
6
wild-type, human CRP transgenic (
CRPtg
), thromboxane receptor–deficient (
Tp
−/−
), and CRPtgTp
−/−
mice. CRP and TP expression were correlated, and suppression of CRP expression using small interfering RNA/CRP led to reduction in TP expression. Platelet–endothelial adherence was increased in CRPtg and suppressed in CRPtgTP
−/−
and CRPtg cells that were suppressed with TP small interfering RNA. TP deficiency in both platelets and endothelial cells was synergistic in affecting platelet–endothelial interactions. Time until arterial occlusion, measured after photochemical injury, was significantly shorter in CRPtg and prolonged in CRPtgTp
−/−
compared with controls (n=10–15, 35±3.4, 136±13.8, and 67±8.9 minutes, respectively;
P
<0.05).
Conclusion—
TP pathway is of major importance in CRP-induced thrombosis. The expression of TP is increased in CRPtg endothelial cells, and its blockade significantly suppresses the prothrombotic effect of CRP.
Collapse
Affiliation(s)
- Etty Grad
- From the Cardiovascular Research Center, Hadassah-Hebrew University Medical Center, Jerusalem, Israel (E.G, R.M.P., H.D.D.); and the Institute for Translational Medicine and Therapeutics, The University of Pennsylvania, Philadelphia, PA (G.A.F.)
| | - Rachel M. Pachino
- From the Cardiovascular Research Center, Hadassah-Hebrew University Medical Center, Jerusalem, Israel (E.G, R.M.P., H.D.D.); and the Institute for Translational Medicine and Therapeutics, The University of Pennsylvania, Philadelphia, PA (G.A.F.)
| | - Garret A. FitzGerald
- From the Cardiovascular Research Center, Hadassah-Hebrew University Medical Center, Jerusalem, Israel (E.G, R.M.P., H.D.D.); and the Institute for Translational Medicine and Therapeutics, The University of Pennsylvania, Philadelphia, PA (G.A.F.)
| | - Haim D. Danenberg
- From the Cardiovascular Research Center, Hadassah-Hebrew University Medical Center, Jerusalem, Israel (E.G, R.M.P., H.D.D.); and the Institute for Translational Medicine and Therapeutics, The University of Pennsylvania, Philadelphia, PA (G.A.F.)
| |
Collapse
|
29
|
Xu X, Liu C, Xu Z, Tzan K, Wang A, Rajagopalan S, Sun Q. Altered adipocyte progenitor population and adipose-related gene profile in adipose tissue by long-term high-fat diet in mice. Life Sci 2012; 90:1001-9. [PMID: 22683431 DOI: 10.1016/j.lfs.2012.05.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Revised: 05/02/2012] [Accepted: 05/18/2012] [Indexed: 12/29/2022]
Abstract
AIMS High-fat diet (HFD) is associated with adipose inflammation, which contributes to key components of metabolic abnormalities. The expanded adipose tissue mass associated with obesity is the result of hyperplasia and hypertrophy of adipocytes. In this study, we investigated the effects of long-term HFD on adipocyte progenitor cell (APC) population and adipose-specific gene profiles in both white and brown adipose, and the role of perivascular adipose in the alteration of vascular function in response to HFD. MAIN METHODS Male C57BL/6 mice were fed a standard normal diet (ND) or HFD for about 8 months. Glucose metabolism was assessed by an intraperitoneal glucose tolerance test. APC population and adipose-related gene profile were evaluated, and vascular function was measured in the presence or absence of perivascular adipose. Adiponectin and AMPK activity were also investigated. KEY FINDINGS HFD induced insulin resistance and glucose intolerance, and resulted in a decrease in APC population in brown, but not in white adipose tissue, when compared with animals fed a ND, with differential alterations of white and brown adipocyte-specific gene expression in brown and white adipose. Additionally, HFD led to altered vascular function in arteries in the presence of perivascular adipose tissue, which is associated with increased superoxide production. Adiponectin and AMPK activity were significantly decreased in response to long-term HFD. SIGNIFICANCE These findings suggest that long-term high-fat intake differentially alters adipocyte progenitor population and adipose-related gene expression in adipose tissue, and adiponectin-AMPK signaling might be involved. In addition, HFD induces changes in perivascular adipose-mediated vascular function.
Collapse
Affiliation(s)
- Xiaohua Xu
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, OH 43210, United States
| | | | | | | | | | | | | |
Collapse
|
30
|
The vasorelaxant mechanisms of a Rho kinase inhibitor DL0805 in rat thoracic aorta. Molecules 2012; 17:5935-44. [PMID: 22609784 PMCID: PMC6268074 DOI: 10.3390/molecules17055935] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 05/11/2012] [Accepted: 05/11/2012] [Indexed: 11/21/2022] Open
Abstract
Rho-kinase has been suggested as a potential therapeutic target in the treatment of cardiovascular diseases. The Rho-kinase signaling pathway is substantially involved in vascular contraction. The aim of the present study was to evaluate the vasorelaxant effects of Rho kinase inhibitor DL0805 in isolated rat aortic rings and to investigate its possible mechanism(s). It was found that DL0805 exerted vasorelaxation in a dose-dependent manner in NE or KCl-induced sustained contraction and partial loss of the vasorelaxation under endothelium-denuded rings. The DL0805-induced vasorelaxation was significantly reduced by the nitric oxide synthase inhibitor Nω-nitro-L-arginine methyl ester, the guanylate cyclase inhibitor methylene blue and the cyclooxygenase inhibitor indomethacin. The voltage-dependent K+ channel blocker 4-aminopyridine remarkably attenuated DL0805-induced relaxations. However, the ATP-sensitive K+ channel blocker glibenclamide and Ca2+-activated K+ channel blocker tetraethylammonium did not affect the DL0805-induced relaxation. In the endothelium-denuded rings, DL0805 also reduced NE-induced transient contraction and inhibited contraction induced by increasing external calcium. These findings suggested that DL0805 is a novel vasorelaxant compound associated with inhibition of Rho/ROCK signaling pathway. The NO-cGMP pathway may be involved in the relaxation of DL0805 in endothelium-intact aorta. The vasorelaxant effect of DL0805 is partially mediated by the opening of the voltage-dependent K+ channels.
Collapse
|
31
|
Félétou M, Huang Y, Vanhoutte PM. Endothelium-mediated control of vascular tone: COX-1 and COX-2 products. Br J Pharmacol 2012; 164:894-912. [PMID: 21323907 DOI: 10.1111/j.1476-5381.2011.01276.x] [Citation(s) in RCA: 262] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Endothelium-dependent contractions contribute to endothelial dysfunction in various animal models of aging, diabetes and cardiovascular diseases. In the spontaneously hypertensive rat, the archetypal model for endothelium-dependent contractions, the production of the endothelium-derived contractile factors (EDCF) involves an increase in endothelial intracellular calcium concentration, the production of reactive oxygen species, the predominant activation of cyclooxygenase-1 (COX-1) and to a lesser extent that of COX-2, the diffusion of EDCF towards the smooth muscle cells and the subsequent stimulation of their thromboxane A2-endoperoxide TP receptors. Endothelium-dependent contractions are also observed in various models of hypertension, aging and diabetes. They generally also involve the generation of COX-1- and/or COX-2-derived products and the activation of smooth muscle TP receptors. Depending on the model, thromboxane A(2), PGH(2), PGF(2α), PGE(2) and paradoxically PGI(2) can all act as EDCFs. In human, the production of COX-derived EDCF is a characteristic of the aging and diseased blood vessels, with essential hypertension causing an earlier onset and an acceleration of this endothelial dysfunction. As it has been observed in animal models, COX-1, COX-2 or both isoforms can contribute to these endothelial dysfunctions. Since in most cases, the activation of TP receptors is the common downstream effector, selective antagonists of this receptor should curtail endothelial dysfunction and be of therapeutic interest in the treatment of cardiovascular disorders.
Collapse
|
32
|
Bone morphogenic protein-4 induces endothelial cell apoptosis through oxidative stress-dependent p38MAPK and JNK pathway. J Mol Cell Cardiol 2011; 52:237-44. [PMID: 22064324 DOI: 10.1016/j.yjmcc.2011.10.013] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 09/16/2011] [Accepted: 10/17/2011] [Indexed: 12/22/2022]
Abstract
The expression of bone morphogenic protein 4 (BMP4), a new pro-inflammatory marker, is increased by disturbed flow in endothelial cells (ECs). BMP4 stimulates production of reactive oxygen species (ROS) and causes endothelial cell dysfunction. The present study examined BMP4-induced apoptosis in ECs and isolated arteries from rat, mouse, and human, and the signaling pathways mediating BMP4-induced apoptosis. Apoptosis was assessed by flow cytometry to detect Annexin-V positive cells, and terminal deoxynucleotidyl transferase dUTP nick end (TUNEL) labeling. The superoxide production was measured by dihydroethidium fluorescence. BMP4 induced EC apoptosis in human mesenteric arteries, mouse aortic endothelium, rat primary ECs, and human ECs. BMP4-induced EC apoptosis was mediated through ROS production by activation of NADPH oxidase, which led to cleaved caspase-3 expression. BMP4 also induced sequential activation of p38 MAPK and JNK which was upstream of caspase 3 activation. Knockdown of BMP receptor 1A by lentiviral shRNA or NOX4 siRNA transfection inhibited BMP4-induced ROS production, p38 and JNK phosphorylation, and caspase-3 activation in ECs. JNK siRNA inhibited BMP4-induced JNK phosphorylation and caspase-3 activation. The present study delineates that BMP4 causes EC apoptosis through activation of caspase-3 in a ROS/p38MAPK/JNK-dependent signaling cascade.
Collapse
|
33
|
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: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
34
|
|
35
|
Yuen CY, Wong WT, Tian XY, Wong SL, Lau CW, Yu J, Tomlinson B, Yao X, Huang Y. Telmisartan inhibits vasoconstriction via PPARγ-dependent expression and activation of endothelial nitric oxide synthase. Cardiovasc Res 2010; 90:122-9. [PMID: 21156825 DOI: 10.1093/cvr/cvq392] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
AIMS Telmisartan activates peroxisome proliferator-activated receptor-γ (PPARγ) in addition to serving as an angiotensin II type 1 receptor (AT(1)R) blocker. The PPARγ activity of telmisartan on resistance arteries has remained largely unknown. The present study investigated the hypothesis that telmisartan inhibited vascular tension in mouse mesenteric resistance arteries, which was attributed to an increased nitric oxide (NO) production through the PPARγ-dependent augmentation of expression and activity of endothelial nitric oxide synthase (eNOS). METHODS AND RESULTS Second-order mesenteric arteries were isolated from male C57BL/6J, eNOS knockout and PPARγ knockout mice and changes in vascular tension were determined by isometric force measurement with a myograph. Expression and activation of relevant proteins were analysed by Western blotting. Real-time NO production was measured by confocal microscopy using the dye DAF. Telmisartan inhibited 9,11-dideoxy-11α,9α-epoxymethanoprostaglandin F(2α) (U46619)- or endothelin-1-induced contractions. An NOS inhibitor, N(G)-nitro-L-arginine methyl ester (l-NAME), or an inhibitor of soluble guanylate cyclase, 1H-[1,2,4]-oxadizolo[4,3-a]quinoxalin-1-one (ODQ), prevented telmisartan-induced inhibition of U46619 contractions. A PPARγ antagonist, GW9662, abolished telmisartan-induced inhibition. Likewise, the PPARγ antagonist rosiglitazone attenuated U46619-induced contractions. The effects of telmisartan and rosiglitazone were prevented by actinomycin-D, a transcription inhibitor. In contrast, losartan, olmesartan, and irbesartan did not inhibit contractions. The inhibition was absent in mesenteric arteries from eNOS knockout or PPARγ knockout mice. Telmisartan augmented eNOS expression, phosphorylation, and NO production, which were reversed by the co-treatment with GW9662. CONCLUSIONS The present results suggest that telmisartan-induced inhibition of vasoconstriction in resistance arteries is mediated through a PPARγ-dependent increase in eNOS expression and activity that is unrelated to AT₁R blockade.
Collapse
Affiliation(s)
- Chi Yung Yuen
- Institute of Vascular Medicine, Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
The stimulation of thromboxane/endoperoxide receptors (TP) elicits diverse physiological/pathophysiological reactions, including platelet aggregation and contraction of vascular smooth muscle. Furthermore, the activation of endothelial TP promotes the expression of adhesion molecules and favors adhesion and infiltration of monocytes/macrophages. In various cardiovascular diseases, endothelial dysfunction is predominantly the result of the release of endothelium-derived contracting factors that counteract the vasodilator effect of nitric oxide produced by the endothelial nitric oxide synthase. Endothelium-dependent contractions involve the activation of cyclooxygenases, the production of reactive oxygen species along with that of endothelium-derived contracting factors, which diffuse toward the vascular smooth muscle cells and activate their TP. TP antagonists curtail the endothelial dysfunction in diseases such as hypertension and diabetes, are potent antithrombotic agents, and reduce vascular inflammation. Therefore, TP antagonists, because of this triple activity, may have a unique potential for the treatment of cardiovascular disorders.
Collapse
|
37
|
Félétou M, Huang Y, Vanhoutte PM. Vasoconstrictor prostanoids. Pflugers Arch 2010; 459:941-50. [PMID: 20333529 DOI: 10.1007/s00424-010-0812-6] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Revised: 02/15/2010] [Accepted: 02/18/2010] [Indexed: 01/20/2023]
Abstract
In cardiovascular diseases and during aging, endothelial dysfunction is due in part to the release of endothelium-derived contracting factors that counteract the vasodilator effect of the nitric oxide. Endothelium-dependent contractions involve the activation of endothelial cyclooxygenases and the release of various prostanoids, which activate thromboxane prostanoid (TP) receptors of the underlying vascular smooth muscle. The stimulation of TP receptors elicits not only the contraction and the proliferation of vascular smooth muscle cells but also diverse physiological/pathophysiological reactions, including platelet aggregation and activation of endothelial inflammatory responses. TP receptor antagonists curtail endothelial dysfunction in diseases such as hypertension and diabetes, are potent antithrombotic agents, and prevent vascular inflammation.
Collapse
|
38
|
Liu CQ, Wong SL, Leung FP, Tian XY, Lau CW, Lu L, Yao X, Chen ZY, Yao T, Huang Y. Prostanoid TP receptor-mediated impairment of cyclic AMP-dependent vasorelaxation is reversed by phosphodiesterase inhibitors. Eur J Pharmacol 2010; 632:45-51. [PMID: 20096281 DOI: 10.1016/j.ejphar.2010.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Revised: 12/18/2009] [Accepted: 01/12/2010] [Indexed: 11/18/2022]
Abstract
Activation of the thromboxane prostanoid (TP) receptor produces potent vasoconstriction, which contributes to the increased vascular tone and blood pressure. The present study was designed to examine the hypothesis that stimulation of prostanoid TP receptors impairs endothelium-independent relaxations to cyclic AMP-elevating agents via increasing the activity of phosphodiesterases (PDEs). Rat carotid arteries without endothelium were isolated and suspended in myograph for the measurement of changes in isometric tension; the tissue content of cyclic AMP was assayed by enzyme immunoassay kit; and prostanoid TP receptor was detected in vascular wall by immunohistochemistry and Western blot. In phenylephrine-contracted rings without endothelium, relaxations induced by isoprenaline (receptor-mediated) and forskolin (receptor-independent) were markedly reduced by the presence of a prostanoid TP receptor agonist, U46619; the attenuated relaxations were prevented by acute treatment with S18886, the selective prostanoid TP receptor antagonist, but not by protein kinase C inhibitors. The reduced relaxations were partially restored by IBMX (non-selective PDE inhibitor), cilostazol (PDE3 inhibitor), rolipram (PDE4 inhibitor) or by Y27632 (Rho kinase inhibitor), but not by T0156 (PDE5 inhibitor). U46619 diminished isoprenaline- or forskolin-stimulated rise in cyclic AMP and this effect was inhibited by cilostazol, rolipram or Y27632. The present results suggest that activation of prostanoid TP receptors impairs cyclic AMP-dependent vasorelaxations partly via PDE- and RhoA/Rho kinase-dependent mechanisms. Inhibitors of PDEs and Rho kinase may be useful in the treatment of cardiovascular complications.
Collapse
Affiliation(s)
- Cui Qing Liu
- Department of Physiology, Hangzhou Normal University, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Cohen RA, Feletou M, Vanhoutte PM, Verbeuren TJ. TP receptors and oxidative stress hand in hand from endothelial dysfunction to atherosclerosis. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2010; 60:85-106. [PMID: 21081216 PMCID: PMC3004095 DOI: 10.1016/b978-0-12-385061-4.00004-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Thromboxane A(2) and the activation of TP receptors that it causes play an important role in platelet aggregation and therefore in thrombosis. However, TP receptors are also involved in the pathologies of the vascular wall including impaired endothelium-dependent vasodilation, increased oxidant generation, and increased expression of adhesion molecules. The beneficial effects of TP antagonists on the vascular wall attenuate these features of vascular disease. They are not shared by aspirin. In fact, TP antagonists are active in patients treated with aspirin, indicating that their potential beneficial effects are mediated by mechanisms different from the antithrombotic actions of aspirin. Our studies have demonstrated the vascular benefits of TP antagonists in experimental animals, particularly in models of diabetes mellitus, in which elevated levels of eicosanoids play a role not only in vascular pathologies but also in those of the kidney and other tissues. They suggest that TP blockade protects against fundamental and widespread tissular dysfunction associated with metabolic disease including hyperlipidemia and hyperglycemia. TP receptor antagonists represent a promising avenue for the prevention of vascular disease in part because of these pleiotropic actions that extend beyond their antithrombotic properties.
Collapse
Affiliation(s)
- Richard A. Cohen
- Vascular Biology Unit, Whitaker Cardiovascular Institute, Boston University School of Medicine
| | - Michel Feletou
- Department of Angiology, Institut de Recherches Servier, Suresnes, France
| | - Paul M. Vanhoutte
- Department Pharmacology and Pharmacy, Li Ka Shing Faculty Medicine, University of Hong Kong, Hong Kong, China and Department BIN Fusion Technology, Chonbuk National University, Jeonju, Korea
| | - Tony J. Verbeuren
- Department of Angiology, Institut de Recherches Servier, Suresnes, France,Correspondence to: Dr Tony J. Verbeuren, Department of Angiology, Institut de Recherches Servier, 11 rue des Moulineaux, Suresnes, France., Tel:
| |
Collapse
|
40
|
Wong SL, Wong WT, Tian XY, Lau CW, Huang Y. Prostaglandins in action indispensable roles of cyclooxygenase-1 and -2 in endothelium-dependent contractions. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2010; 60:61-83. [PMID: 21081215 DOI: 10.1016/b978-0-12-385061-4.00003-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Endothelium regulates local vascular tone by means of releasing relaxing and contracting factors, of which the latter have been found to be elevated in vascular pathogenesis of hypertension, diabetes, hypercholesterolemia, and aging. Endothelium-derived contracting factors (EDCFs) are mainly metabolites of arachidonic acid generated by cyclooxygenase (COX), as vasodilatations in patients with hypertension, metabolic diseases, or advancing age are improved by acute treatment with COX inhibitor indomethacin. COX is presented in two isoforms, COX-1 and COX-2, with the former regarded as constitutive and the latter mainly expressed upon induction. Experiments with animal models of vascular dysfunctions, however, reveal that both isoforms have similar capacity to participate in endothelium-dependent contractions, with augmented expression and activity. COX-derived prostaglandin (PG) H(2), PGF(2α), PGE(2), prostacyclin (PGI(2)), and thromboxane A(2) (TxA(2)) are the proposed EDCFs that mediate endothelium-dependent contractions via the activation of thromboxane-prostanoid (TP) receptor in various vascular beds from different species. Although COX inhibition seems to be a possible strategy in combating COX-associated vascular complications, the incidence of adverse cardiovascular effects of Vioxx has greatly antagonized this concept. Further review of COX inhibitors is required, especially toward the selectivity of coxibs and whether it directly inhibits prostacyclin synthase activity. Meanwhile, TP receptor antagonism may emerge as a therapeutic alternative to reverse prostanoid-mediated vascular dysregulations.
Collapse
Affiliation(s)
- Siu Ling Wong
- Institute of Vascular Medicine, Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | | | | | | | | |
Collapse
|