Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cell Signal ; 107: 110676, 2023 07.
Article in English | MEDLINE | ID: mdl-37028778

ABSTRACT

Asymmetric dimethylarginine (ADMA) is generated through the irreversible methylation of arginine residues. It is an independent risk factor for cardiovascular disease, currently thought to be due to its ability to act as a competitive inhibitor of the nitric oxide (NO) synthase enzymes. Plasma ADMA concentrations increase with obesity and fall following weight loss; however, it is unknown whether they play an active role in adipose pathology. Here, we demonstrate that ADMA drives lipid accumulation through a newly identified NO-independent pathway via the amino-acid sensitive calcium-sensing receptor (CaSR). ADMA treatment of 3T3-L1 and HepG2 cells upregulates a suite of lipogenic genes with an associated increase in triglyceride content. Pharmacological activation of CaSR mimics ADMA while negative modulation of CaSR inhibits ADMA driven lipid accumulation. Further investigation using CaSR overexpressing HEK293 cells demonstrated that ADMA potentiates CaSR signalling via Gq intracellular Ca2+ mobilisation. This study identifies a signalling mechanism for ADMA as an endogenous ligand of the G protein-coupled receptor CaSR that potentially contributes to the impact of ADMA in cardiometabolic disease.


Subject(s)
Arginine , Receptors, Calcium-Sensing , Humans , HEK293 Cells , Arginine/metabolism , Nitric Oxide Synthase/metabolism , Lipids
2.
Circ Res ; 127(6): 747-760, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32539601

ABSTRACT

RATIONALE: Metabolic syndrome (MetS) is a cluster of interrelated risk factors for cardiovascular diseases and atherosclerosis. Circulating levels of large extracellular vesicles (lEVs), submicrometer-sized vesicles released from plasma membrane, from MetS patients were shown to induce endothelial dysfunction, but their role in early stage of atherosclerosis and on vascular smooth muscle cells (SMC) remain to be fully elucidated. OBJECTIVE: To determine the mechanisms by which lEVs lead to the progression of atherosclerosis in the setting of MetS. METHODS AND RESULTS: Proteomic analysis revealed that the small GTPase, Rap1 was overexpressed in lEVs from MetS patients compared with those from non-MetS subjects. Rap1 was in GTP-associated active state in both types of lEVs, and Rap1-lEVs levels correlated with increased cardiovascular risks, including stenosis. MetS-lEVs, but not non-MetS-lEVs, increased Rap1-dependent endothelial cell permeability. MetS-lEVs significantly promoted migration and proliferation of human aortic SMC and increased expression of proinflammatory molecules and activation of ERK (extracellular signal-regulated kinase) 5/p38 pathways. Neutralization of Rap1 by specific antibody or pharmacological inhibition of Rap1 completely prevented the effects of lEVs from MetS patients. High-fat diet-fed ApoE-/- mice displayed an increased expression of Rap1 both in aortas and circulating lEVs. lEVs accumulated in plaque atherosclerotic lesions depending on the progression of atherosclerosis. lEVs from high-fat diet-fed ApoE-/- mice, but not those from mice fed with a standard diet, enhanced SMC proliferation. Human atherosclerotic lesions were enriched in lEVs expressing Rap1. CONCLUSIONS: These data demonstrate that Rap1 carried by MetS-lEVs participates in the enhanced SMC proliferation, migration, proinflammatory profile, and activation of ERK5/p38 pathways leading to vascular inflammation and remodeling, and atherosclerosis. These results highlight that Rap1 carried by MetS-lEVs may be a novel determinant of diagnostic value for cardiometabolic risk factors and suggest Rap1 as a promising therapeutic target against the development of atherosclerosis. Graphical Abstract: A graphical abstract is available for this article.


Subject(s)
Atherosclerosis/metabolism , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic , rap1 GTP-Binding Proteins/metabolism , Adult , Aged , Aged, 80 and over , Animals , Atherosclerosis/blood , Atherosclerosis/pathology , Case-Control Studies , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Female , Humans , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Middle Aged , Mitogen-Activated Protein Kinase 7/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Permeability , Phosphorylation , Prognosis , Proteomics , Risk Assessment , Risk Factors , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , rap GTP-Binding Proteins
3.
Front Pharmacol ; 9: 406, 2018.
Article in English | MEDLINE | ID: mdl-29740325

ABSTRACT

Red wine polyphenol extracts improve cardiovascular and metabolic disorders linked to obesity. Their vascular protection is mediated by the activation of the alpha isoform of the estrogen receptor (ERα). In the present study, we explored the effects of a grape seed extract (GSE) enriched in the flavan-3-ols procyanidin dimers on obesity-related cardiovascular and metabolic disorders; with a particular interest in the role/contribution of ERα. Ovariectomized wild type or ERα knockout (KO) mice were fed with standard or western diet, supplemented or not with GSE, for 12 weeks. Their body weight was monitored throughout the study, and an echocardiography was performed at the end of the treatment. Blood and tissues were collected for biochemical and functional analysis, including nitric oxide and oxidative stress measurement. Vascular reactivity and liver mitochondrial complexes activity were also analyzed. In western diet-fed mice, GSE reduced adiposity, plasma triglycerides, and oxidative stress in the heart, liver, adipose and skeletal tissues; but did not improve the vascular dysfunction. In western diet-fed mice, ERα deletion prevented or reduced the beneficial effects of GSE on plasma triglycerides and visceral adiposity. ERα deletion also prevented/reduced the anti-oxidant effect of GSE in the liver, but did not affect its capacity to reduce oxidative stress in the heart and adipose tissue. In conclusion, dietary supplementation of GSE attenuated features of metabolic syndrome partially through ERα-dependent mechanisms. This report highlights the therapeutic potential of polyphenols, and especially extract enriched in procyanidin dimers, against the metabolic disorders associated with excessive energy intake.

4.
Cardiovasc Res ; 113(3): 276-287, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28395021

ABSTRACT

Aims: RhoB plays a key role in the pathogenesis of hypoxia-induced pulmonary hypertension. Farnesylated RhoB promotes growth responses in cancer cells and we investigated whether inhibition of protein farnesylation will have a protective effect. Methods and results: The analysis of lung tissues from rodent models and pulmonary hypertensive patients showed increased levels of protein farnesylation. Oral farnesyltransferase inhibitor tipifarnib prevented development of hypoxia-induced pulmonary hypertension in mice. Tipifarnib reduced hypoxia-induced vascular cell proliferation, increased endothelium-dependent vasodilatation and reduced vasoconstriction of intrapulmonary arteries without affecting cell viability. Protective effects of tipifarnib were associated with inhibition of Ras and RhoB, actin depolymerization and increased eNOS expression in vitro and in vivo. Farnesylated-only RhoB (F-RhoB) increased proliferative responses in cultured pulmonary vascular cells, mimicking the effects of hypoxia, while both geranylgeranylated-only RhoB (GG-RhoB), and tipifarnib had an inhibitory effect. Label-free proteomics linked F-RhoB with cell survival, activation of cell cycle and mitochondrial biogenesis. Hypoxia increased and tipifarnib reduced the levels of F-RhoB-regulated proteins in the lung, reinforcing the importance of RhoB as a signalling mediator. Unlike simvastatin, tipifarnib did not increase the expression levels of Rho proteins. Conclusions: Our study demonstrates the importance of protein farnesylation in pulmonary vascular remodelling and provides a rationale for selective targeting of this pathway in pulmonary hypertension.


Subject(s)
Antihypertensive Agents/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Hypertension, Pulmonary/prevention & control , Hypoxia/drug therapy , Pulmonary Artery/drug effects , Quinolones/pharmacology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/pathology , Farnesyltranstransferase/metabolism , Humans , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/etiology , Hypoxia/complications , Hypoxia/enzymology , Male , Mice, Inbred C57BL , Phenotype , Protein Prenylation , Proteomics/methods , Pulmonary Artery/enzymology , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Time Factors , Transfection , Vasoconstriction/drug effects , Vasodilation/drug effects , rhoB GTP-Binding Protein/genetics , rhoB GTP-Binding Protein/metabolism
5.
Arterioscler Thromb Vasc Biol ; 36(10): 2078-87, 2016 10.
Article in English | MEDLINE | ID: mdl-27470511

ABSTRACT

OBJECTIVE: Inflammation and dysregulated angiogenesis are features of endothelial dysfunction in pulmonary hypertension. Neutrophil extracellular traps (NETs), produced by dying neutrophils, contribute to pathogenesis of numerous vascular disorders but their role in pulmonary hypertension has not been studied. We sought evidence of (NETs) formation in pulmonary hypertension and investigated the effect of NETs on endothelial function. APPROACH AND RESULTS: Plasma and lung tissues of patients with pulmonary hypertension were analyzed for NET markers. The effects of NETs on endothelial function were studied in vitro and in vivo. Patients with chronic thromboembolic pulmonary hypertension and idiopathic pulmonary hypertension showed elevated plasma levels of DNA, neutrophil elastase, and myeloperoxidase. NET-forming neutrophils and extensive areas of NETosis were found in the occlusive plexiform lesions and vascularized intrapulmonary thrombi. NETs induced nuclear factor κB-dependent endothelial angiogenesis in vitro and increased vascularization of matrigel plugs in vivo. Angiogenic responses were associated with increased release of matrix metalloproteinase-9, heparin-binding epidermal growth factor-like growth factor, latency-associated peptide of the transforming growth factor ß1, and urokinase-type plasminogen activator, accompanied by increased endothelial permeability and cell motility. NETs-induced responses depended on myeloperoxidase/H2O2-dependent activation of Toll-like receptor 4/nuclear factor κB signaling. NETs stimulated the release of endothelin-1 in HPAECs (human pulmonary artery endothelial cells) and stimulated pulmonary smooth muscle cell proliferation in vitro. CONCLUSIONS: We are the first to implicate NETs in angiogenesis and provide a functional link between NETs and inflammatory angiogenesis in vitro and in vivo. We demonstrate the potential pathological relevance of this in 2 diseases of disordered vascular homeostasis, pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension.


Subject(s)
Endothelial Cells/metabolism , Extracellular Traps/metabolism , Hypertension, Pulmonary/metabolism , Neovascularization, Pathologic , Neutrophils/metabolism , Pulmonary Artery/metabolism , Animals , Case-Control Studies , Cell Movement , Cell Proliferation , Cells, Cultured , Chloride Channels/genetics , Chloride Channels/metabolism , Coculture Techniques , Endothelial Cells/pathology , Humans , Hydrogen Peroxide/metabolism , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Male , Mice , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Neutrophils/pathology , Peroxidase/metabolism , Pulmonary Artery/pathology , RNA Interference , Signal Transduction , Toll-Like Receptor 4/metabolism , Transfection
6.
Exp Eye Res ; 147: 148-155, 2016 06.
Article in English | MEDLINE | ID: mdl-27181226

ABSTRACT

Ischemia-induced angiogenesis is critical for tissue repair, but aberrant neovascularization in the retina causes severe sight impairment. Nitric oxide (NO) has been implicated in neovascular eye disease because of its pro-angiogenic properties in the retina. Nitric oxide production is inhibited endogenously by asymmetric dimethylarginines (ADMA and L-NMMA) which are metabolized by dimethylarginine dimethylaminohydrolase (DDAH) 1 and 2. The aim of this study was to determine the roles of DDAH1, DDAH2, ADMA and L-NMMA in retinal ischemia-induced angiogenesis. First, DDAH1, DDAH2, ADMA and L-NMMA levels were determined in adult C57BL/6J mice. The results obtained revealed that DDAH1 was twofold increased in the retina compared to the brain and the choroid. DDAH2 expression was approximately 150 fold greater in retinal and 70 fold greater in choroidal tissue compared to brain tissue suggesting an important tissue-specific role for DDAH2 in the retina and choroid. ADMA and L-NMMA levels were similar in the retina and choroid under physiological conditions. Next, characterization of DDAH1(+/-) and DDAH2(-/-) deficient mice by in vivo fluorescein angiography, immunohistochemistry and electroretinography revealed normal neurovascular function compared with wildtype control mice. Finally, DDAH1(+/-) and DDAH2(-/-) deficient mice were studied in the oxygen-induced retinopathy (OIR) model, a model used to emulate retinal ischemia and neovascularization, and VEGF and ADMA levels were quantified by ELISA and liquid chromatography tandem mass spectrometry. In the OIR model, DDAH1(+/-) exhibited a similar phenotype compared to wildtype controls. DDAH2 deficiency, in contrast, resulted in elevated retinal ADMA which was associated with attenuated aberrant angiogenesis and improved vascular regeneration in a VEGF independent manner. Taken together this study suggests, that in retinal ischemia, DDAH2 deficiency elevates ADMA, promotes vascular regeneration and protects against aberrant angiogenesis. Therapeutic inhibition of DDAH2 may therefore offer a potential therapeutic strategy to protect sight by promoting retinal vascular regeneration and preventing pathological angiogenesis.


Subject(s)
Amidohydrolases/deficiency , Choroidal Neovascularization/metabolism , Retinal Neovascularization/metabolism , Amidohydrolases/metabolism , Animals , Choroidal Neovascularization/physiopathology , Disease Models, Animal , Electroretinography , Immunohistochemistry , Ischemia/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/metabolism , Nitric Oxide/metabolism , Retinal Neovascularization/physiopathology , Vascular Endothelial Growth Factor A/metabolism
7.
J Cardiovasc Transl Res ; 9(2): 162-4, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26960567

ABSTRACT

Pulmonary arterial hypertension (PAH) is a chronic and progressive disease which continues to carry an unacceptably high mortality and morbidity. The nitric oxide (NO) pathway has been implicated in the pathophysiology and progression of the disease. Its extremely short half-life and systemic effects have hampered the clinical use of NO in PAH. In an attempt to circumvent these major limitations, we have developed a new NO-nanomedicine formulation. The formulation was based on hydrogel-like polymeric composite NO-releasing nanoparticles (NO-RP). The kinetics of NO release from the NO-RP showed a peak at about 120 min followed by a sustained release for over 8 h. The NO-RP did not affect the viability or inflammation responses of endothelial cells. The NO-RP produced concentration-dependent relaxations of pulmonary arteries in mice with PAH induced by hypoxia. In conclusion, NO-RP drugs could considerably enhance the therapeutic potential of NO therapy for PAH.


Subject(s)
Antihypertensive Agents/pharmacology , Arterial Pressure/drug effects , Hypertension, Pulmonary/drug therapy , Nanoparticles , Nitric Oxide Donors/pharmacology , Nitric Oxide/metabolism , Pulmonary Artery/drug effects , Animals , Antihypertensive Agents/chemistry , Antihypertensive Agents/metabolism , Dose-Response Relationship, Drug , Drug Compounding , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Kinetics , Mice , Nanomedicine , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/metabolism , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology
8.
Front Pharmacol ; 7: 529, 2016.
Article in English | MEDLINE | ID: mdl-28119607

ABSTRACT

Red wine polyphenol extracts (polyphenols) ameliorate cardiovascular and metabolic disorders associated with obesity. Previously, we demonstrated that the alpha isoform of estrogen receptor (ERα) triggers the vascular protection of polyphenols. Here, we investigated the contribution of ERα on the effects of polyphenols on cardiovascular and metabolic alterations associated with obesity. We used ovariectomized wild type or ERα-deficient mice receiving standard (SD) or western (WD) diets, or SD and WD containing polyphenols (SD+polyphenols and WD+polyphenols, respectively) over a 12-week period. Body weight was measured during treatment. Echocardiography examination was performed before sacrifice. Blood and tissues were sampled for biochemical and functional analysis with respect to nitric oxide (NO•) and oxidative stress. Vascular reactivity and liver mitochondrial complexes were analyzed. In WD-fed mice, polyphenols reduced adiposity, plasma triglycerides and oxidative stress in aorta, heart, adipose and liver tissues and enhanced NO• production in aorta and liver. ERα deletion prevented or reduced the beneficial effects of polyphenols, especially visceral adiposity, aortic and liver oxidative stresses and NO• bioavailability. ERα deletion, however, had no effect on polyphenol's ability to decrease the fat accumulation and oxidative stress of subcutaneous adipose tissue. Also, ERα deletion did not modify the decrease of ROS levels induced by polyphenols treatment in the visceral adipose tissue and heart from WD-fed mice. Dietary supplementation of polyphenols remarkably attenuates features of metabolic syndrome; these effects are partially mediated by ERα-dependent mechanisms. This study demonstrates the therapeutic potential of this extract in metabolic and cardiovascular alterations linked to excessive energy intake.

9.
Biochem Pharmacol ; 92(4): 607-17, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25451690

ABSTRACT

Growing evidence suggests that hepatic-insulin resistance is sufficient to promote progression to cardiovascular disease. We have shown previously that liver-specific protein-tyrosine-phosphatase 1B (PTP1B) deficiency improves hepatic-insulin sensitivity and whole-body glucose homeostasis. The aim of this study was to investigate the impact of liver-specific PTP1B-deficiency (L-PTP1B-/-) on cardiac and peripheral vascular function, with special emphasis on endothelial function in the context of high-fat diet (HFD)-induced obesity. L-PTP1B-/- mice exhibited an improved glucose and lipid homeostasis and increased insulin sensitivity, without changes in body weight. HFD-feeding increased systolic blood pressure (BP) in both L-PTP1B-/- and control littermates; however, this was significantly lower in L-PTP1B-/- mice. HFD-feeding increased diastolic BP in control mice only, whilst the L-PTP1B-/- mice were completely protected. The analysis of the function of the left ventricle (LV) revealed that HFD-feeding decreased LV fractional shortening in control animals, which was not observed in L-PTP1B-/- mice. Importantly, HFD feeding significantly impaired endothelium-dependent vasorelaxation in response to acetylcholine in aortas from control mice, whilst L-PTP1B-/- mice were fully protected. This was associated with alterations in eNOS phosphorylation. Selective inhibition of COX-2, using NS-398, decreased the contractile response in response to serotonin (5-HT) only in vessels from control mice. HFD-fed control mice released enhanced levels of prostaglandin E, a vasoconstrictor metabolite; whilst both chow- and HFD-fed L-PTP1B-/- mice released higher levels of prostacylin, a vasorelaxant metabolite. Our data indicate that hepatic-PTP1B inhibition protects against HFD-induced endothelial dysfunction, underscoring the potential of peripheral PTP1B inhibitors in reduction of obesity-associated cardiovascular risk in addition to its anti-diabetic effects.


Subject(s)
Endothelium, Vascular/physiopathology , Liver/enzymology , Obesity/physiopathology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Animals , Blood Pressure , Electrocardiography , Endothelium, Vascular/enzymology , Glucose/metabolism , Homeostasis , Lipid Metabolism , Mice , Mice, Knockout , Obesity/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics
10.
Biochem J ; 462(1): 103-12, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24895913

ABSTRACT

The NOS (nitric oxide synthase) inhibitor ADMA (asymmetric dimethylarginine) contributes to the pathogenesis of pulmonary hypertension. Reduced levels of the enzymes metabolizing ADMA, dimethylarginine dimethylaminohydrolases (DDAH1 and DDAH2) and increased levels of miR-21 are linked to disease pathology, but the mechanisms are not understood. In the present study we assessed the potential role of miR-21 in the regulation of hypoxia-induced changes in ADMA metabolism in vitro and in vivo. Hypoxia inhibited DDAH1 and DDAH2 expression and increased ADMA levels in cultured human pulmonary endothelial cells. In contrast, in human pulmonary smooth muscle cells, only DDAH2 was reduced whereas ADMA levels remained unchanged. Endothelium-specific down-regulation of DDAH1 by miR-21 in hypoxia induced endothelial dysfunction and was prevented by overexpression of DDAH1 and miR-21 blockade. DDAH1, but not DDAH2, mRNA levels were reduced, whereas miR-21 levels were elevated in lung tissues from patients with pulmonary arterial hypertension and mice with pulmonary hypertension exposed to 2 weeks of hypoxia. Hypoxic mice treated with miR-21 inhibitors and DDAH1 transgenic mice showed elevated lung DDAH1, increased cGMP levels and attenuated pulmonary hypertension. Regulation of DDAH1 by miR-21 plays a role in the development of hypoxia-induced pulmonary hypertension and may be of broader significance in pulmonary hypertension.


Subject(s)
Amidohydrolases/metabolism , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , MicroRNAs/physiology , Animals , Arginine/analogs & derivatives , Cells, Cultured , Cyclic GMP/metabolism , Endothelial Cells/metabolism , Familial Primary Pulmonary Hypertension , Humans , Hypertension, Pulmonary/genetics , Lung/blood supply , Lung/metabolism , Male , Mice , Mice, Transgenic , Myocytes, Smooth Muscle/metabolism
11.
Int J Biochem Cell Biol ; 53: 9-14, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24792670

ABSTRACT

Delphinidin, an anthocyanin present in red wine, has been reported to exert vasculoprotective properties on endothelial cells, including vasorelaxing and anti-apoptotic effects. Moreover, delphinidin treatment in a rat model of post-ischemic neovascularization has been described to exert anti-angiogenic property. Angiogenesis is an energetic process and VEGF-induced angiogenesis is associated with mitochondrial biogenesis. However, whether delphinidin induces changes in mitochondrial biogenesis has never been addressed. Effects of delphinidin were investigated in human endothelial cells at a concentration described to be anti-angiogenic in vitro (10(-2)g/l). mRNA expression of mitochondrial biogenesis factors, mitochondrial respiration, DNA content and enzyme activities were assessed after 48 h of stimulation. Delphinidin increased mRNA expression of several mitochondrial biogenesis factors, including NRF1, ERRα, Tfam, Tfb2m and PolG but did not affect neither mitochondrial respiration, DNA content nor enzyme activities. In presence of delphinidin, VEGF failed to increase mitochondrial respiration, DNA content, complex IV activity and Akt activation in endothelial cells. These results suggest a possible association between inhibition of VEGF-induced mitochondrial biogenesis through Akt pathway by delphinidin and its anti-angiogenic effect, providing a novel mechanism sustaining the beneficial effect of delphinidin against pathologies associated with excessive angiogenesis such as cancers.


Subject(s)
Anthocyanins/administration & dosage , Cell Respiration/genetics , Neoplasms/genetics , Proto-Oncogene Proteins c-akt/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis , Angiogenesis Inhibitors/administration & dosage , Animals , Apoptosis/drug effects , Cell Respiration/drug effects , Endothelial Cells/drug effects , Humans , Mitochondrial Turnover/genetics , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Proto-Oncogene Proteins c-akt/genetics , Rats , Signal Transduction/drug effects
12.
Cell Tissue Res ; 355(3): 675-85, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24599334

ABSTRACT

Pulmonary endothelial permeability is an important determinant of vascular adaptation to changes in oxygen tension, blood pressure, levels of growth factors or inflammatory cytokines. The Ras homologous (Rho) family of guanosine triphosphate phosphatases (Rho GTPases), key regulators of the actin cytoskeleton, regulate endothelial barrier function in response to a variety of environmental factors and signalling agents via the reorganization of the actin cytoskeleton, changes in receptor trafficking or the phosphorylation of junctional proteins. This review provides a brief summary of recent knowledge on Rho-GTPase-mediated effects on pulmonary endothelial barrier function and focuses in particular on their role in pulmonary vascular disorders, including pulmonary hypertension, chronic obstructive pulmonary disease, acute lung injury and acute respiratory distress syndrome.


Subject(s)
Actins/physiology , Capillary Permeability/physiology , Endothelium, Vascular/physiology , Lung/physiology , rho GTP-Binding Proteins/physiology , Actins/metabolism , Animals , Endothelium, Vascular/metabolism , Humans , Lung/cytology , Lung/enzymology , rho GTP-Binding Proteins/metabolism
13.
PLoS One ; 8(11): e82594, 2013.
Article in English | MEDLINE | ID: mdl-24312428

ABSTRACT

Tetrahydrobiopterin (BH4), which fosters the formation of and stabilizes endothelial NO synthase (eNOS) as an active dimer, tightly regulates eNOS coupling / uncoupling. Moreover, studies conducted in genetically-modified models demonstrate that BH4 pulmonary deficiency is a key determinant in the pathogenesis of pulmonary hypertension. The present study thus investigates biopterin metabolism and eNOS expression, as well as the effect of sepiapterin (a precursor of BH4) and eNOS gene deletion, in a mice model of hypoxic pulmonary hypertension. In lungs, chronic hypoxia increased BH4 levels and eNOS expression, without modifying dihydrobiopterin (BH2, the oxidation product of BH4) levels, GTP cyclohydrolase-1 or dihydrofolate reductase expression (two key enzymes regulating BH4 availability). In intrapulmonary arteries, chronic hypoxia also increased expression of eNOS, but did not induce destabilisation of eNOS dimers into monomers. In hypoxic mice, sepiapterin prevented increase in right ventricular systolic pressure and right ventricular hypertrophy, whereas it modified neither remodelling nor alteration in vasomotor responses (hyper-responsiveness to phenylephrine, decrease in endothelium-dependent relaxation to acetylcholine) in intrapulmonary arteries. Finally, deletion of eNOS gene partially prevented hypoxia-induced increase in right ventricular systolic pressure, right ventricular hypertrophy and remodelling of intrapulmonary arteries. Collectively, these data demonstrate the absence of BH4/BH2 changes and eNOS dimer destabilisation, which may induce eNOS uncoupling during hypoxia-induced pulmonary hypertension. Thus, even though eNOS gene deletion and sepiapterin treatment exert protective effects on hypoxia-induced pulmonary vascular remodelling, increase on right ventricular pressure and / or right ventricular hypertrophy, these effects appear unrelated to biopterin-dependent eNOS uncoupling within pulmonary vasculature of hypoxic wild-type mice.


Subject(s)
Biopterins/metabolism , Hypertension, Pulmonary/metabolism , Hypoxia/complications , Nitric Oxide Synthase Type III/metabolism , Animals , Disease Models, Animal , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/enzymology , Mice , Nitric Oxide Synthase Type III/genetics , Tetrahydrofolate Dehydrogenase/metabolism
14.
Int J Biochem Cell Biol ; 45(4): 783-91, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23333619

ABSTRACT

Red wine polyphenolic compounds (RWPC) are reported to exert vasculoprotective properties on endothelial cells, involving nitric oxide (NO) release via a redox-sensitive pathway. This NO release involves the activation of the estrogen receptor-alpha (ERα). Paradoxical effects of a RWPC treatment occur in a rat model of post-ischemic neovascularization, where a low-dose is pro-angiogenic while a higher dose is anti-angiogenic. NO and ERα are key regulators of mitochondrial capacity, and angiogenesis is a highly energetic process associated with mitochondrial biogenesis. However, whether RWPC induces changes in mitochondrial capacity has never been addressed. We investigated the effects of RWPC at low (10(-4)g/l, LCP) and high concentration (10(-2)g/l, HCP) in human endothelial cells. Mitochondrial respiration, expression of mitochondrial biogenesis factors and mitochondrial DNA content were assessed using oxygraphy and quantitative PCR respectively. In vitro capillary formation using ECM gel(®) was also performed. Treatment with LCP increased mitochondrial respiration, with a maximal effect achieved at 48h. LCP also increased expression of several mitochondrial biogenesis factors and mitochondrial DNA content. In contrast, HCP did not affect these parameters. Furthermore, LCP modulated both mitochondrial capacity and angiogenesis through mechanisms sensitive to ER, NADPH oxidase and NO-synthase inhibitors. Finally, the inhibition of mitochondrial protein synthesis abolished the pro-angiogenic capacity of LCP. These results suggest a possible association between the modulation of mitochondrial capacity by LCP and its pro-angiogenic activity. These data provide evidence for a role of mitochondria in the regulation of angiogenesis by RWPC.


Subject(s)
Mitochondria/drug effects , NADPH Oxidases/metabolism , Neovascularization, Physiologic/drug effects , Nitric Oxide Synthase/metabolism , Polyphenols/pharmacology , Receptors, Estrogen/metabolism , Wine , Acetophenones/pharmacology , Cell Respiration/drug effects , Chloramphenicol/pharmacology , DNA, Mitochondrial/metabolism , Dose-Response Relationship, Drug , Estradiol/analogs & derivatives , Estradiol/pharmacology , Fulvestrant , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mitochondria/metabolism , NADPH Oxidases/antagonists & inhibitors , Nitric Oxide Synthase/antagonists & inhibitors , Nitroarginine/pharmacology , Phenols/pharmacology , Pyrazoles/pharmacology , Receptors, Estrogen/agonists , Receptors, Estrogen/antagonists & inhibitors , Signal Transduction/drug effects
15.
Clin Sci (Lond) ; 123(3): 173-92, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22494160

ABSTRACT

Understanding the role of oxidative injury will allow for therapy with agents that scavenge ROS (reactive oxygen species) and antioxidants in the management of several diseases related to free radical damage. The majority of free radicals are generated by mitochondria as a consequence of the mitochondrial cycle, whereas free radical accumulation is limited by the action of a variety of antioxidant processes that reside in every cell. In the present review, we provide an overview of the mitochondrial generation of ROS and discuss the role of ROS in the regulation of endothelial and adipocyte function. Moreover, we also discuss recent findings on the role of ROS in sepsis, cerebral ataxia and stroke. These results provide avenues for the therapeutic potential of antioxidants in a variety of diseases.


Subject(s)
Antioxidants/metabolism , Systems Biology , Animals , Humans , Mitochondria/metabolism , Models, Biological , Oxygen/metabolism , Reactive Oxygen Species/metabolism
16.
PLoS One ; 7(3): e34268, 2012.
Article in English | MEDLINE | ID: mdl-22457831

ABSTRACT

AIMS: Obesity is a primary contributor to acquired insulin resistance leading to the development of type 2 diabetes and cardiovascular alterations. The carnitine derivate, propionyl-L-carnitine (PLC), plays a key role in energy control. Our aim was to evaluate metabolic and cardiovascular effects of PLC in diet-induced obese mice. METHODS: C57BL/6 mice were fed a high-fat diet for 9 weeks and then divided into two groups, receiving either free- (vehicle-HF) or PLC-supplemented water (200 mg/kg/day) during 4 additional weeks. Standard diet-fed animals were used as lean controls (vehicle-ST). Body weight and food intake were monitored. Glucose and insulin tolerance tests were assessed, as well as the HOMA(IR), the serum lipid profile, the hepatic and muscular mitochondrial activity and the tissue nitric oxide (NO) liberation. Systolic blood pressure, cardiac and endothelial functions were also evaluated. RESULTS: Vehicle-HF displayed a greater increase of body weight compared to vehicle-ST that was completely reversed by PLC treatment without affecting food intake. PLC improved the insulin-resistant state and reversed the increased total cholesterol but not the increase in free fatty acid, triglyceride and HDL/LDL ratio induced by high-fat diet. Vehicle-HF exhibited a reduced cardiac output/body weight ratio, endothelial dysfunction and tissue decrease of NO production, all of them being improved by PLC treatment. Finally, the decrease of hepatic mitochondrial activity by high-fat diet was reversed by PLC. CONCLUSIONS: Oral administration of PLC improves the insulin-resistant state developed by obese animals and decreases the cardiovascular risk associated to this metabolic alteration probably via correction of mitochondrial function.


Subject(s)
Carnitine/analogs & derivatives , Diet , Electron Transport/drug effects , Liver/drug effects , Animals , Body Weight/drug effects , Carnitine/pharmacology , Feeding Behavior/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Organ Size/drug effects
17.
PLoS One ; 6(11): e27809, 2011.
Article in English | MEDLINE | ID: mdl-22110764

ABSTRACT

Microparticles are membrane vesicles with pro-inflammatory properties. Circulating levels of microparticles have previously been found to be elevated in patients with metabolic syndrome (MetS). The present study aimed to evaluate the effects of in vivo treatment with microparticles, from patients with MetS and from healthy subjects (HS), on ex vivo vascular function in mice. Microparticles isolated from MetS patients or HS, or a vehicle were intravenously injected into mice, following which vascular reactivity in response to vasoconstrictor agonists was assessed by myography with respect to cyclo-oxygenase pathway, oxidative and nitrosative stress. Injection of microparticles from MetS patients into mice induced vascular hypo-reactivity in response to serotonin. Hypo-reactivity was associated with up-regulation of inducible NO-synthase and increased production of NO, and was reversed by the NO-synthase inhibitor (N(G)-nitro-L-arginine). The selective COX-2 inhibitor (NS398) reduced the contractile effect of serotonin in aortas from mice treated with vehicle or HS microparticles; however, this was not observed within mice treated with MetS microparticles, probably due to the ability of MetS microparticles to enhance prostacyclin. MetS microparticle-mediated vascular dysfunction was associated with increased reactive oxygen species (ROS) and enhanced expression of the NADPH oxidase subunits. Neutralization of the pro-inflammatory pathway Fas/FasL completely prevented vascular hypo-reactivity and the ability of MetS microparticles to enhance both inducible NO-synthase and monocyte chemoattractant protein-1 (MCP-1). Our data provide evidence that microparticles from MetS patients induce ex vivo vascular dysfunction by increasing both ROS and NO release and by altering cyclo-oxygenase metabolites and MCP-1 through the Fas/FasL pathway.


Subject(s)
Aorta/cytology , Aorta/metabolism , Cell-Derived Microparticles/metabolism , Fas Ligand Protein/metabolism , Metabolic Syndrome/pathology , Signal Transduction , fas Receptor/metabolism , Animals , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cyclooxygenase 2/metabolism , Female , Gene Expression Regulation, Enzymologic , Humans , Male , Mice , Middle Aged , NADPH Oxidases/metabolism , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism
18.
J. physiol. biochem ; 67(3): 285-296, sept. 2011.
Article in English | IBECS | ID: ibc-122594

ABSTRACT

No disponible


Mitochondria have been shown to be impaired in insulin resistance-related diseases but have not been extensively studied during the first steps of adipose cell development. This study was designed to determine the sequence of changes of the mitochondrial network and function during the first days of adipogenesis. 3T3-L1 preadipocytes were differentiated into adipocytes without using glitazone compounds. At days 0, 3, 6, 9, and 12, mitochondrial network imaging, mitochondrial oxygen consumption, membrane potential, and oxidative phosphorylation efficiency were assessed in permeabilized cells. Gene and protein expressions related to fatty acid metabolism and mitochondrial network were also determined. Compared to preadipocytes (day 0), new adipocytes (days 6 and 9) displayed profound (..) (AU)


Subject(s)
Humans , Cell Differentiation/physiology , Mitochondria/physiology , 3T3-L1 Cells/physiology , Oxidative Stress/physiology , Insulin Resistance/physiology , Adipogenesis/physiology
19.
J Physiol Biochem ; 67(3): 285-96, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21267801

ABSTRACT

Mitochondria have been shown to be impaired in insulin resistance-related diseases but have not been extensively studied during the first steps of adipose cell development. This study was designed to determine the sequence of changes of the mitochondrial network and function during the first days of adipogenesis. 3T3-L1 preadipocytes were differentiated into adipocytes without using glitazone compounds. At days 0, 3, 6, 9, and 12, mitochondrial network imaging, mitochondrial oxygen consumption, membrane potential, and oxidative phosphorylation efficiency were assessed in permeabilized cells. Gene and protein expressions related to fatty acid metabolism and mitochondrial network were also determined. Compared to preadipocytes (day 0), new adipocytes (days 6 and 9) displayed profound changes of their mitochondrial network that underwent fragmentation and redistribution around lipid droplets. Drp1 and mitofusin 2 displayed a progressive increase in their gene expression and protein content during the first 9 days of differentiation. In parallel with the mitochondrial network redistribution, mitochondria switched to uncoupled respiration with a tendency towards decreased membrane potential, with no variation of mtTFA and NRF1 gene expression. The expression of PGC1α and NRF2 genes and genes involved in lipid oxidation (UCP2, CD36, and CPT1) was increased. Reactive oxygen species (ROS) production displayed a nadir at day 6 with a concomitant increase in antioxidant enzyme gene expression. This 3T3-L1-based in vitro model of adipogenesis showed that mitochondria adapted to the increased number of lipid droplets by network redistribution and uncoupling respiration. The timing and regulation of lipid oxidation-associated ROS production appeared to play an important role in these changes.


Subject(s)
Adipocytes/physiology , Cell Differentiation , Mitochondria/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adipogenesis , Adiponectin/metabolism , Animals , Catalase/genetics , Catalase/metabolism , Citrate (si)-Synthase/metabolism , Electron Transport Complex IV/metabolism , Enzyme Assays , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Gene Expression , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , L-Lactate Dehydrogenase/metabolism , Membrane Potential, Mitochondrial , Mice , Microscopy, Video , Mitochondria/enzymology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nitric Oxide/metabolism , Oxidation-Reduction , Oxygen Consumption , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...