Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mater Sci Eng C Mater Biol Appl ; 112: 110938, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32409084

RESUMO

Fe-based materials are currently considered for manufacturing biodegradable coronary stents. Here we show that Fe has a strong potential to generate hydroxyl radicals (HO) during corrosion. This HO generation, but not corrosion, can be inhibited by catalase. Oxidative stress was observed (increased HO-1 expression) in aortic rings after direct exposure to Fe, but not in the presence of catalase or after indirect exposure. This oxidative stress response induced an uncoupling of eNOS in, and a consequent reduced NO production by endothelial cells exposed to Fe. In isolated rat aortic rings NO production was also reduced by HO generated during Fe corrosion, as indicated by the protective role of catalase. Finally, all these mechanisms contributed to impaired endothelium-dependent relaxation in aortic rings caused by HO generated during the direct contact with Fe. This deleterious impact of Fe corrosion on the endothelial function should be integrated when considering the use of biodegradable Fe-based alloys for vascular implants.


Assuntos
Radical Hidroxila/metabolismo , Ferro/química , Stents , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/metabolismo , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Carbacol/farmacologia , Catalase/metabolismo , Bovinos , Corrosão , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Heme Oxigenase-1/metabolismo , Radical Hidroxila/toxicidade , Masculino , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Próteses e Implantes , Ratos , Ratos Wistar
2.
Oncogene ; 35(30): 3976-85, 2016 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-26686091

RESUMO

Photosensitizers (PS) are ideally devoid of any activity in the absence of photoactivation, and rely on molecular oxygen for the formation of singlet oxygen ((1)O2) to produce cellular damage. Off-targets and tumor hypoxia therefore represent obstacles for the use of PS for cancer photodynamic therapy. Herein, we describe the characterization of OR141, a benzophenazine compound identified through a phenotypic screening for its capacity to be strictly activated by light and to kill a large variety of tumor cells under both normoxia and hypoxia. This new class of PS unraveled an unsuspected common mechanism of action for PS that involves the combined inhibition of the mammalian target of rapamycin (mTOR) signaling pathway and proteasomal deubiquitinases (DUBs) USP14 and UCH37. Oxidation of mTOR and other endoplasmic reticulum (ER)-associated proteins drives the early formation of high molecular weight (MW) complexes of multimeric proteins, the concomitant blockade of DUBs preventing their degradation and precipitating cell death. Furthermore, we validated the antitumor effects of OR141 in vivo and documented its highly selective accumulation in the ER, further increasing the ER stress resulting from (1)O2 generation upon light activation.


Assuntos
Enzimas Desubiquitinantes/antagonistas & inibidores , Retículo Endoplasmático/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Oxigênio/farmacologia , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Animais , Hipóxia Celular , Linhagem Celular Tumoral , Humanos , Camundongos , Neoplasias/metabolismo , Oxirredução , Complexo de Endopeptidases do Proteassoma/metabolismo , Serina-Treonina Quinases TOR/fisiologia
3.
Channels (Austin) ; 8(5): 402-13, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25483583

RESUMO

Smooth muscle myosin light chain kinase (MLCK) plays a crucial role in artery contraction, which regulates blood pressure and blood flow distribution. In addition to this role, MLCK contributes to Ca(2+) flux regulation in vascular smooth muscle (VSM) and in non-muscle cells, where cytoskeleton has been suggested to help Ca(2+) channels trafficking. This conclusion is based on the use of pharmacological inhibitors of MLCK and molecular and cellular techniques developed to down-regulate the enzyme. Dissimilarities have been observed between cells and whole tissues, as well as between large conductance and small resistance arteries. A differential expression in MLCK and ion channels (either voltage-dependent Ca(2+) channels or non-selective cationic channels) could account for these observations, and is in line with the functional properties of the arteries. A potential involvement of MLCK in the pathways modulating Ca(2+) entry in VSM is described in the present review.


Assuntos
Canais de Cálcio/metabolismo , Músculo Liso/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Animais , Humanos , Músculo Liso/enzimologia
4.
Pflugers Arch ; 466(7): 1377-89, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24162233

RESUMO

The Ca(2+)-dependent kinase myosin light chain kinase (MLCK) is the activator of smooth muscle contraction. In addition, it has been reported to be involved in Ca(2+) channel regulation in cultured cells, and we previously showed that the MLCK inhibitor ML-7 decreases arginine vasopressin (AVP)-induced Ca(2+) influx in rat aorta. This study was designed to investigate whether MLCK is involved in Ca(2+) regulation in resistance artery smooth muscle cell, which plays a major role in the control of blood pressure. As ML compounds were shown to have off-target effects, MLCK was downregulated by transfection with a small interfering RNA targeting MLCK (MLCK-siRNA) in rat small resistance mesenteric artery (RMA) and in the rat embryonic aortic cell line A7r5. Noradrenaline-induced contraction and Ca(2+) signal were significantly depressed in MLCK-siRNA compared to scramble-siRNA-transfected RMA. Contraction and Ca(2+) signal induced by high KCl and voltage-activated Ca(2+) current were also significantly decreased in MLCK-siRNA-transfected RMA, suggesting that MLCK depletion modifies voltage-operated Ca(2+) channels. KCl- and AVP-induced Ca(2+) signals and voltage-activated Ca(2+) current were decreased in MLCK-depleted A7r5 cells. Eventually, real-time quantitative PCR analysis indicated that in A7r5, MLCK controlled mRNA expression of CaV1.2 (L-type) and CaV3.1 (T-type) voltage-dependent Ca(2+) channels. Our results suggest that MLCK controls the transcription of voltage-dependent Ca(2+) channels in vascular smooth muscle cells.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo T/metabolismo , Músculo Liso Vascular/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Animais , Arginina Vasopressina/farmacologia , Cálcio/metabolismo , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo T/genética , Linhagem Celular , Masculino , Contração Muscular , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Quinase de Cadeia Leve de Miosina/genética , Norepinefrina/farmacologia , Cloreto de Potássio/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Vasoconstrição
5.
Pflugers Arch ; 466(2): 237-51, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23873354

RESUMO

The water channels, aquaporins (AQPs) are key mediators of transcellular fluid transport. However, their expression and role in cardiac tissue is poorly characterized. Particularly, AQP1 was suggested to transport other molecules (nitric oxide (NO), hydrogen peroxide (H2O2)) with potential major bearing on cardiovascular physiology. We therefore examined the expression of all AQPs and the phenotype of AQP1 knockout mice (vs. wild-type littermates) under implanted telemetry in vivo, as well as endothelium-dependent relaxation in isolated aortas and resistance vessels ex vivo. Four aquaporins were expressed in wild-type heart tissue (AQP1, AQP7, AQP4, AQP8) and two aquaporins in aortic and mesenteric vessels (AQP1-AQP7). AQP1 was expressed in endothelial as well as cardiac and vascular muscle cells and co-segregated with caveolin-1. AQP1 knockout (KO) mice exhibited a prominent microcardia and decreased myocyte transverse dimensions despite no change in capillary density. Both male and female AQP1 KO mice had lower mean BP, which was not attributable to altered water balance or autonomic dysfunction (from baroreflex and frequency analysis of BP and HR variability). NO-dependent BP variability was unperturbed. Accordingly, endothelium-derived hyperpolarizing factor (EDH(F)) or NO-dependent relaxation were unchanged in aorta or resistance vessels ex vivo. However, AQP1 KO mesenteric vessels exhibited an increase in endothelial prostanoids-dependent relaxation, together with increased expression of COX-2. This enhanced relaxation was abrogated by COX inhibition. We conclude that AQP1 does not regulate the endothelial EDH or NO-dependent relaxation ex vivo or in vivo, but its deletion decreases baseline BP together with increased prostanoids-dependent relaxation in resistance vessels. Strikingly, this was associated with microcardia, unrelated to perturbed angiogenesis. This may raise interest for new inhibitors of AQP1 and their use to treat hypertrophic cardiac remodeling.


Assuntos
Aquaporina 1/deficiência , Pressão Sanguínea/fisiologia , Animais , Aquaporina 1/fisiologia , Fatores Biológicos/fisiologia , Feminino , Cardiopatias Congênitas/patologia , Hipotensão/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Contração Miocárdica/fisiologia , Óxido Nítrico/fisiologia
6.
Physiol Rev ; 89(2): 481-534, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19342613

RESUMO

Nitric oxide production in response to flow-dependent shear forces applied on the surface of endothelial cells is a fundamental mechanism of regulation of vascular tone, peripheral resistance, and tissue perfusion. This implicates the concerted action of multiple upstream "mechanosensing" molecules reversibly assembled in signalosomes recruiting endothelial nitric oxide synthase (eNOS) in specific subcellular locales, e.g., plasmalemmal caveolae. Subsequent short- and long-term increases in activity and expression of eNOS translate this mechanical stimulus into enhanced NO production and bioactivity through a complex transcriptional and posttranslational regulation of the enzyme, including by shear-stress responsive transcription factors, oxidant stress-dependent regulation of transcript stability, eNOS regulatory phosphorylations, and protein-protein interactions. Notably, eNOS expressed in cardiac myocytes is amenable to a similar regulation in response to stretching of cardiac muscle cells and in part mediates the length-dependent increase in cardiac contraction force. In addition to short-term regulation of contractile tone, eNOS mediates key aspects of cardiac and vascular remodeling, e.g., by orchestrating the mobilization, recruitment, migration, and differentiation of cardiac and vascular progenitor cells, in part by regulating the stabilization and transcriptional activity of hypoxia inducible factor in normoxia and hypoxia. The continuum of the influence of eNOS in cardiovascular biology explains its growing implication in mechanosensitive aspects of integrated physiology, such as the control of blood pressure variability or the modulation of cardiac remodeling in situations of hemodynamic overload.


Assuntos
Fenômenos Fisiológicos Cardiovasculares , Mecanotransdução Celular/fisiologia , Óxido Nítrico Sintase Tipo III/fisiologia , Animais , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Humanos , Contração Miocárdica/fisiologia , Miocárdio/citologia , Vasoconstrição/fisiologia
7.
J Physiol Pharmacol ; 60 Suppl 4: 105-9, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20083858

RESUMO

Endothelium plays a crucial role in the regulation of cardiovascular homeostasis through the release of vasoactive factors. Nitric oxide (NO) and endothelium-derived hyperpolarizing factors (EDHF) are the two major actors controlling the vasomotor tone. The endothelial nitric oxide synthase (eNOS) was reported in the mid 90ies to be under the control of caveolin, the structural protein of caveolae. Nowadays, a large body of evidence has confirmed that the caveolin/eNOS interaction was needed to prevent inadequate NO production under basal conditions but also to facilitate the integration of extracellular stimuli to intracellular NO signals. Compartmentation of key actors in the EDHF signaling pathway is now also proposed to take place into caveolae. Accordingly, caveolin-deficient animals revealed both an unopposed NO production promoting vessel dilation and a lack of EDHF-driven vasorelaxation. The transient receptor potential (TRP) channels are the link between caveolae and EDHF. Different TRP channels involved in the capacitative calcium entry were found to directly interact with caveolin-1 in endothelial cells. TRPC1 and TRPC4 form a complex with the endoplasmic reticulum IP3 receptor thereby optimizing calcium signaling. EDHF-driven vasodilation was documented to be altered in a TRPV4-deficient mouse model. The close vicinity between TRPV4 and SKCa channels in caveolae together with the gap-junctions subunits connexins support a role of these microdomains in the generation and propagation of EDHF to vascular smooth muscle cells. In conclusion, caveolae and caveolin are important control points in the control of blood pressure by the endothelium. This also highlights how any alteration in the caveolae integrity or caveolin abundance may lead to and/or exacerbate endothelial dysfunction and associated cardiovascular diseases.


Assuntos
Fatores Biológicos/fisiologia , Cavéolas/fisiologia , Caveolinas/fisiologia , Músculo Liso Vascular/fisiologia , Óxido Nítrico/fisiologia , Animais , Fatores Biológicos/metabolismo , Humanos , Camundongos , Tono Muscular/fisiologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/fisiologia , Transdução de Sinais/fisiologia
8.
Circulation ; 117(8): 1065-74, 2008 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-18268148

RESUMO

BACKGROUND: In endothelial cells, caveolin-1, the structural protein of caveolae, acts as a scaffolding protein to cluster lipids and signaling molecules within caveolae and, in some instances, regulates the activity of proteins targeted to caveolae. Specifically, different putative mediators of the endothelium-derived hyperpolarizing factor (EDHF)-mediated relaxation are located in caveolae and/or regulated by the structural protein caveolin-1, such as potassium channels, calcium regulatory proteins, and connexin 43, a molecular component of gap junctions. METHODS AND RESULTS: Comparing relaxation in vessels from caveolin-1 knockout mice and their wild-type littermates, we observed a complete absence of EDHF-mediated vasodilation in isolated mesenteric arteries from caveolin-1 knockout mice. The absence of caveolin-1 is associated with an impairment of calcium homeostasis in endothelial cells, notably, a decreased activity of Ca2+-permeable TRPV4 cation channels that participate in nitric oxide- and EDHF-mediated relaxation. Moreover, morphological characterization of caveolin-1 knockout and wild-type arteries showed fewer gap junctions in vessels from knockout animals associated with a lower expression of connexins 37, 40, and 43 and altered myoendothelial communication. Finally, we showed that TRPV4 channels and connexins colocalize with caveolin-1 in the caveolar compartment of the plasma membrane. CONCLUSIONS: We demonstrated that expression of caveolin-1 is required for EDHF-related relaxation by modulating membrane location and activity of TRPV4 channels and connexins, which are both implicated at different steps in the EDHF-signaling pathway.


Assuntos
Fatores Biológicos/metabolismo , Sinalização do Cálcio/fisiologia , Caveolina 1/metabolismo , Compartimento Celular/fisiologia , Células Endoteliais/metabolismo , Vasodilatação/fisiologia , Animais , Cálcio/metabolismo , Cavéolas/metabolismo , Caveolina 1/genética , Conexinas/metabolismo , Células Endoteliais/ultraestrutura , Junções Comunicantes/metabolismo , Camundongos , Camundongos Knockout , Microcirculação , Óxido Nítrico/metabolismo , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo
9.
Eur J Heart Fail ; 9(12): 1163-71, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17999941

RESUMO

OBJECTIVE: To analyze the implication of the beta3-adrenoceptor (beta3-AR) pathway in human septic myocardium and a murine model of sepsis, a condition associated with myocardial depression. METHODS AND RESULTS: beta3-AR and eNOS protein abundance were increased (332+/-66.4% and 218+/-39.3; P<0.05) in hearts from septic patients. The effect of BRL37344, a beta3-AR-preferential agonist, was analyzed by videomicroscopy on the contractility of neonatal mouse ventricular myocytes (NMVM) incubated with conditioned medium from LPS-stimulated cultured macrophages (Mc-LPS+ medium). Stimulation of untreated NMVM with BRL37344 dose-dependently decreased the amplitude of contractile shortening (P<0.05). This response was abolished by L-NAME (NOS inhibitor). Incubation in Mc-LPS+ medium potentiated the depressing effect of BRL37344 (P<0.05) as well as of SR58611A (P<0.05) in wild-type myocytes. Importantly, the contractile depression was abrogated in cardiomyocytes from beta3-AR KO mice. CONCLUSIONS: beta3-AR are upregulated during sepsis in the human myocardium and by cytokines in murine cardiomyocytes, where they mediate an increased negative inotropic response to beta3 agonists. Activation of the beta3-AR pathway by catecholamines may contribute to the myocardial dysfunction in sepsis.


Assuntos
Miócitos Cardíacos/metabolismo , RNA/genética , Receptores Adrenérgicos beta 3/genética , Sepse/metabolismo , Regulação para Cima , Agonistas de Receptores Adrenérgicos beta 3 , Agonistas Adrenérgicos beta/farmacologia , Adulto , Animais , Animais Recém-Nascidos , Western Blotting , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Modelos Animais de Doenças , Etanolaminas/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/fisiologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , RNA/biossíntese , Ratos , Ratos Wistar , Receptores Adrenérgicos beta 3/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sepse/patologia , Sepse/fisiopatologia , Estereoisomerismo
10.
Circulation ; 110(8): 948-54, 2004 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-15302798

RESUMO

BACKGROUND: Coronary vessel tone is modulated in part by beta-adrenergic relaxation. However, the implication of specific beta-adrenoceptor subtypes and their downstream vasorelaxing mechanism(s) in human coronary resistance arteries is poorly defined. beta3-Adrenoceptors were recently shown to vasodilate animal vessels and are expressed in human hearts. METHODS AND RESULTS: We examined the expression and functional role of beta3-adrenoceptors in human coronary microarteries and their coupling to vasodilating nitric oxide (NO) and/or hyperpolarization mechanisms. The expression of beta3-adrenoceptor mRNA and protein was demonstrated in extracts of human coronary microarteries. Immunohistochemical analysis revealed their exclusive localization in the endothelium, with no staining of vascular smooth muscle. In contractility experiments in which videomicroscopy was used, the nonspecific beta-agonist isoproterenol and the beta3-preferential agonist BRL37344 evoked an approximately 50% relaxation of endothelin-1-preconstricted human coronary microarteries. Relaxations were blocked by the beta1/beta2/beta3-adrenoceptor antagonist bupranolol but were insensitive to the beta1/beta2-adrenoceptor antagonist nadolol, confirming a beta3-adrenoceptor-mediated pathway. Relaxation in response to BRL37344 was absent in human coronary microarteries devoid of functional endothelium. When human coronary microarteries were precontracted with KCl (thereby preventing vessel hyperpolarization), the relaxation to BRL37344 was reduced to 15.5% and totally abrogated by the NO synthase inhibitor L-omega-nitroarginine, confirming the participation of a NO synthase-mediated relaxation. The NO synthase-independent relaxation was completely inhibited by the Ca2+-activated K+ channel inhibitors apamin and charybdotoxin, consistent with an additional endothelium-derived hyperpolarizing factor-like response. Accordingly, membrane potential recordings demonstrated vessel hyperpolarization in response to beta3-adrenoceptor stimulation. CONCLUSIONS: Beta3-adrenoceptors are expressed in the endothelium of human coronary resistance arteries and mediate adrenergic vasodilatation through both NO and vessel hyperpolarization.


Assuntos
Vasos Coronários/fisiologia , Endotélio Vascular/fisiologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico/fisiologia , Receptores Adrenérgicos beta 3/fisiologia , Vasodilatação/fisiologia , Adolescente , Agonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Apamina/farmacologia , Arteríolas/efeitos dos fármacos , Arteríolas/fisiologia , Bupranolol/farmacologia , Charibdotoxina/farmacologia , Criança , Vasos Coronários/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Etanolaminas/farmacologia , Feminino , Humanos , Isoproterenol/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Microcirculação/efeitos dos fármacos , Microcirculação/fisiologia , Microscopia de Vídeo , Pessoa de Meia-Idade , Nadolol/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III , Nitroarginina/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , RNA Mensageiro/biossíntese , Receptores Adrenérgicos beta 1/efeitos dos fármacos , Receptores Adrenérgicos beta 2/efeitos dos fármacos , Receptores Adrenérgicos beta 3/biossíntese , Receptores Adrenérgicos beta 3/efeitos dos fármacos , Receptores Adrenérgicos beta 3/genética , Vasodilatação/efeitos dos fármacos
11.
Circ Res ; 93(5): 388-98, 2003 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-12958142

RESUMO

Nitric oxide (NO) is produced from virtually all cell types composing the myocardium and regulates cardiac function through both vascular-dependent and -independent effects. The former include regulation of coronary vessel tone, thrombogenicity, and proliferative and inflammatory properties as well as cellular cross-talk supporting angiogenesis. The latter comprise the direct effects of NO on several aspects of cardiomyocyte contractility, from the fine regulation of excitation-contraction coupling to modulation of (presynaptic and postsynaptic) autonomic signaling and mitochondrial respiration. This multifaceted involvement of NO in cardiac physiology is supported by a tight molecular regulation of the three NO synthases, from cellular spatial confinement to posttranslational allosteric modulation by specific interacting proteins, acting in concert to restrict the influence of NO to a particular intracellular target in a stimulus-specific manner. Loss of this specificity, such as produced on excessive NO delivery from inflammatory cells (or cytokine-stimulated cardiomyocytes themselves), may result in profound cellular disturbances leading to heart failure. Future therapeutic manipulations of cardiac NO synthesis will necessarily draw on additional characterization of the cellular and molecular determinants for the net effect of this versatile radical on the cardiomyocyte biology.


Assuntos
Coração/fisiologia , Óxido Nítrico/metabolismo , Animais , Coração/fisiopatologia , Cardiopatias/fisiopatologia , Humanos , Contração Miocárdica/fisiologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo I
12.
Am J Physiol Heart Circ Physiol ; 282(1): H219-31, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11748066

RESUMO

Because nitric oxide (NO) regulates cardiac and vessel contraction, we compared the expression and activity of the endothelial NO synthase (eNOS) and caveolin, which tonically inhibits eNOS in normal and hypertrophic cardiomyopathic hearts. NOS activity (L-[(3)H]citrulline formation), eNOS immunostaining, and caveolin abundance were measured in heart tissue of 23 mongrel dogs before and at 3 and 7 wk of perinephritic hypertension (PHT). Hemodynamic parameters in vivo and endothelial NO-dependent relaxation of macro- and coronary microvessels in vitro were assessed in the same animals. eNOS immunostaining and total calcium-dependent NOS activity decreased at 7 wk in all four heart cavities (in left ventricle, from 17.0 +/- 1.3 to 0.2 +/- 0.2 fmol. min(-1). mg protein(-1), P < 0.001). Caveolin-1 and -3 also decreased in PHT dog hearts. Accordingly, basal vascular tone was preserved, but maximal endothelial NO-dependent relaxation was impaired in all vessels from 7-wk PHT dogs. The latter had preserved systolic function but impaired diastolic relaxation [relaxation time constant (T(1)), 25.1 +/- 0.9 vs. 22.0 +/- 1 ms in controls; P < 0.05]. Peripheral infusion of the NOS inhibitor N(G)-nitro-L-arginine methyl ester increased mean aortic pressure in both groups and reduced diastolic (T(1), 31.9 +/- 1.4 ms) and systolic function in PHT dogs (DP40, 47.5 +/- 2.5 vs. 59.4 +/- 3.8 s(-1) in control animals). In conclusion, both eNOS and caveolin proteins are decreased in the hypertrophic hearts of PHT dogs. This is associated with altered maximal (but not basal) vascular relaxation and impaired diastolic function. Further degradation of cardiac function after NOS inhibition suggests a critical role of residual NOS activity, probably supported by the concurrent downregulation of caveolin.


Assuntos
Cardiomiopatia Hipertrófica/fisiopatologia , Caveolinas/metabolismo , Hemodinâmica , Miocárdio/enzimologia , Óxido Nítrico Sintase/metabolismo , Animais , Cardiomiopatia Hipertrófica/patologia , Caveolina 1 , Circulação Coronária/fisiologia , Modelos Animais de Doenças , Cães , Ecocardiografia , Hipertensão Renal/fisiopatologia , Immunoblotting , Imuno-Histoquímica , Artérias Mesentéricas/fisiopatologia , Microcirculação/efeitos dos fármacos , Microcirculação/fisiologia , Contração Muscular , Miocárdio/patologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase Tipo III , Nitroarginina/farmacologia , Análise de Regressão , Função Ventricular Esquerda/fisiologia
13.
Circ Res ; 89(10): 866-73, 2001 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-11701613

RESUMO

3-Hydroxy-3-methylglutaryl (HMG)-coenzyme A reductase inhibitors or statins exert direct beneficial effects on the endothelium in part through an increase in nitric oxide (NO) production. Here, we examined whether posttranslational modifications of the endothelial NO synthase (eNOS) could account for the proangiogenic effects of statins. We used endothelial cells (ECs) isolated from cardiac microvasculature, aorta, and umbilical veins, as well as dissected microvessels and aortic rings, that were cultured on reconstituted basement membrane matrix (Matrigel). Tube or precapillary formation was evaluated after statin treatment, in parallel with immunoblotting and immunoprecipitation experiments. Atorvastatin stimulated NO-dependent angiogenesis from both isolated and outgrowing (vessel-derived) ECs, independently of changes in eNOS expression. We found that in macro- but not microvascular ECs, atorvastatin stabilized tube formation through a decrease in caveolin abundance and its inhibitory interaction with eNOS. We also identified the chaperone protein hsp90 as a key target for the proangiogenic effects of statins. Using geldanamycin, an inhibitor of hsp90 function, and overexpression of recombinant hsp90, we documented that the statin-induced phosphorylation of eNOS on Ser1177 was directly dependent on the ability of hsp90 to recruit Akt in the eNOS complex. Finally, we showed that statin promoted the tyrosine phosphorylation of hsp90 and the direct interaction of hsp90 with Akt, which further potentiated the NO-dependent angiogenic processes. Our study provides new mechanistic insights into the NO-mediated angiogenic effects of statins and underscores the potential of these drugs and other modulators of hsp90 and caveolin abundance to promote neovascularization in disease states associated or not with atherosclerosis.


Assuntos
Caveolinas/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Óxido Nítrico/metabolismo , Proteínas Serina-Treonina Quinases , Animais , Atorvastatina , Benzoquinonas , Bioensaio/métodos , Bovinos , Caveolina 1 , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Inibidores Enzimáticos/farmacologia , Expressão Gênica , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/genética , Ácidos Heptanoicos/farmacologia , Humanos , Immunoblotting , Lactamas Macrocíclicas , Camundongos , Modelos Cardiovasculares , Óxido Nítrico Sintase/efeitos dos fármacos , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Pirróis/farmacologia , Quinonas/farmacologia , Ratos , Transfecção
14.
Nat Cell Biol ; 3(10): 867-73, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11584267

RESUMO

Stretching of cardiac muscle modulates contraction through the enhancement of the Ca2+ transient, but how this occurs is still not known. We found that stretching of myocytes modulates the elementary Ca2+ release process from ryanodine-receptor Ca2+-release channels (RyRCs), Ca2+ sparks and the electrically stimulated Ca2+ transient. Stretching induces PtdIns-3-OH kinase (PI(3)K)-dependent phosphorylation of both Akt and the endothelial isoform of nitric oxide synthase (NOS), nitric oxide (NO) production, and a proportionate increase in Ca2+-spark frequency that is abolished by inhibiting NOS and PI(3)K. Exogenously generated NO reversibly increases Ca2+-spark frequency without cell stretching. We propose that myocyte NO produced by activation of the PI(3)K-Akt-endothelial NOS axis acts as a second messenger of stretch by enhancing RyRC activity, contributing to myocardial contractile activation.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Contração Miocárdica/fisiologia , Miocárdio/metabolismo , Óxido Nítrico/metabolismo , Proteínas Serina-Treonina Quinases , Animais , Estimulação Elétrica , Inibidores Enzimáticos/farmacologia , Corantes Fluorescentes/metabolismo , Masculino , Contração Miocárdica/efeitos dos fármacos , Miocárdio/citologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo III , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Ratos Sprague-Dawley , S-Nitroso-N-Acetilpenicilamina/farmacologia , Espectrometria de Fluorescência
15.
J Biol Chem ; 276(35): 32663-9, 2001 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-11425855

RESUMO

Vascular endothelial growth factor (VEGF) exerts its angiogenic effects partly through the activation of endothelial nitric-oxide synthase (eNOS). Association with heat shock protein 90 (hsp90) and phosphorylation by Akt were recently shown to separately activate eNOS upon VEGF stimulation in endothelial cells. Here, we examined the interplay between these different mechanisms in VEGF-exposed endothelial cells. We documented that hsp90 binding to eNOS is, in fact, the crucial event triggering the transition from the Ca(2+)-dependent activation of eNOS to the phosphorylation-mediated potentiation of its activity by VEGF. Accordingly, we showed that early VEGF stimulation first leads to the Ca(2+)/calmodulin disruption of the caveolin-eNOS complex and promotes the association between eNOS and hsp90. eNOS-bound hsp90 can then recruit VEGF-activated (phosphorylated) Akt to the complex, which in turn can phosphorylate eNOS. Further experiments in transfected COS cells expressing either wild-type or S1177A mutant eNOS led us to identify the serine 1177 as the critical residue for the hsp90-dependent Akt-mediated activation of eNOS. Finally, we documented that although the VEGF-induced phosphorylation of eNOS leads to a sustained production of NO independently of a maintained increase in [Ca(2+)](i), this late stage of eNOS activation is strictly conditional on the initial VEGF-induced Ca(2+)-dependent stimulation of the enzyme. These data establish the critical temporal sequence of events leading to the sustained activation of eNOS by VEGF and suggest new ways of regulating the production of NO in response to this cytokine through the ubiquitous chaperone protein, hsp90.


Assuntos
Cálcio/metabolismo , Fatores de Crescimento Endotelial/farmacologia , Endotélio Vascular/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Linfocinas/farmacologia , Óxido Nítrico Sintase/metabolismo , Proteínas Serina-Treonina Quinases , Animais , Animais Recém-Nascidos , Células COS , Calcimicina/farmacologia , Caveolina 1 , Caveolinas/metabolismo , Células Cultivadas , Chlorocebus aethiops , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Ativação Enzimática , Proteínas de Choque Térmico HSP90/isolamento & purificação , Humanos , Insulina/farmacologia , Cinética , Camundongos , Óxido Nítrico Sintase/isolamento & purificação , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Nitritos/metabolismo , Fosforilação , Ligação Proteica , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteínas Recombinantes/metabolismo , Transfecção , Veias Umbilicais , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
16.
Am J Vet Res ; 62(6): 889-94, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11400846

RESUMO

OBJECTIVE: To assess morphologic and metabolic abnormalities in dogs with early left ventricular dysfunction (ELVD) induced by rapid right ventricular pacing (RRVP). ANIMALS: 7 Beagles. PROCEDURE: Plasma carnitine concentrations were measured before and after development of ELVD induced by RRVP. At the same times, transvenous endomyocardial biopsy was performed, and specimens were submitted for determination of myocardial carnitine concentrations and histologic, morphometric, and ultrastructural examination. RESULTS: In 4 dogs in which baseline plasma total carnitine concentration was normal, RRVP induced a decrease in myocardial total and free carnitine concentrations and an increase in myocardial esterified carnitine concentration. In 3 dogs in which baseline plasma total carnitine concentration was low, plasma and myocardial carnitine concentrations were unchanged after pacing. Structural changes associated with pacing included perinuclear vacuolization in 3 dogs. Morphometric analyses indicated there was a decrease in myofiber cross-sectional diameter and area following pacing. Electron microscopy revealed changes in myofibrils and mitochondria following pacing. CONCLUSIONS AND CLINICAL RELEVANCE: Results indicated that moderate to severe alterations in myocyte cytoarchitecture are present in dogs with ELVD induced by RRVP and that in dogs with normal plasma carnitine concentrations, myocardial carnitine deficiency may be a biochemical marker of ELVD. Results also indicated that transvenous endomyocardial biopsy can be used to evaluate biochemical and structural myocardial changes in dogs with cardiac disease.


Assuntos
Carnitina/metabolismo , Doenças do Cão/metabolismo , Miocárdio/metabolismo , Disfunção Ventricular Esquerda/veterinária , Animais , Biópsia/veterinária , Carnitina/sangue , Doenças do Cão/etiologia , Doenças do Cão/patologia , Cães , Histocitoquímica/veterinária , Masculino , Microscopia Eletrônica/veterinária , Miocárdio/patologia , Miocárdio/ultraestrutura , Taquicardia/etiologia , Taquicardia/patologia , Taquicardia/veterinária , Disfunção Ventricular Esquerda/etiologia , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/patologia
17.
Am J Physiol Heart Circ Physiol ; 280(3): H1422-6, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11179093

RESUMO

The total unbound calmodulin (i.e., not bound to target proteins) level in living smooth muscle cells from the ferret portal vein was monitored with a low-affinity, calmodulin-binding peptide tagged with an environmentally sensitive fluorophore. GS17C, a previously characterized peptide, from the calmodulin-binding domain of caldesmon was tagged with iodoacetyl nitrobenz-2-oxa-1,3-diazole (NBD) or, as a negative control, with iodoacetylfluorescein isothiocyanate. Increases in NBD-GS17C fluorescence were detected by using confocal microscopy when chemically loaded cells were stimulated with solutions of elevated [K(+)] or the calcium ionophore 4-bromoA-23187 to elicit increases in intracellular Ca(2+) concentration ([Ca(2+)](i)) quantified by fura 2. Increases in peptide fluorescence were detected in response to a phorbol ester in the absence of changes in [Ca(2+)](i). These changes were blocked by the addition of the calmodulin antagonist calmidazolium. These results suggest that the total unbound intracellular calmodulin levels may be sufficient to regulate the activity of caldesmon and, furthermore, that phosphorylation of protein kinase C substrates may increase the level of available calmodulin in living smooth muscle cells.


Assuntos
Cálcio/metabolismo , Calmodulina/metabolismo , Músculo Liso Vascular/enzimologia , Proteínas Quinases/metabolismo , 4-Cloro-7-nitrobenzofurazano , Animais , Proteínas de Ligação a Calmodulina/metabolismo , Furões , Indicadores e Reagentes , Músculo Liso Vascular/citologia , Veia Porta/citologia , Veia Porta/metabolismo
18.
Circulation ; 103(1): 113-8, 2001 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-11136695

RESUMO

BACKGROUND: Hypercholesterolemia is causally associated with defects of endothelial nitric oxide (NO)-dependent vasodilation. Increased uptake of cholesterol by endothelial cells (ECs) upregulates the abundance of the structural protein caveolin-1 and impairs NO release through the stabilization of the inhibitory heterocomplex between caveolin-1 and endothelial NO synthase (eNOS). Therefore, we examined whether the hydroxy-methylglutaryl-coenzyme A reductase inhibitor atorvastatin modulates caveolin abundance, eNOS activity, and NO release through a reduction in endogenous cholesterol levels. METHODS AND RESULTS: ECs were incubated with increasing doses of atorvastatin in the absence or in the presence of human LDL cholesterol (LDL-Chol) fractions in the presence of antioxidants. Our results show that atorvastatin (10 nmol/L to 1 micromol/L) reduced caveolin-1 abundance in the absence (-75%) and in the presence (-20% to 70%) of LDL-Chol. This was paralleled by a decreased inhibitory interaction between caveolin-1 and eNOS and a restoration and/or potentiation of the basal (+45%) and agonist-stimulated (+107%) eNOS activity. These effects were observed in the absence of changes in eNOS abundance and were reversed with mevalonate. In the presence of LDL-Chol, atorvastatin also promoted the agonist-induced association of eNOS and the chaperone Hsp90, resulting in the potentiation of eNOS activation. CONCLUSIONS: We provide biochemical and functional evidence that atorvastatin promotes NO production by decreasing caveolin-1 expression in ECs, regardless of the level of extracellular LDL-Chol. These findings highlight the therapeutic potential of inhibiting cholesterol synthesis in peripheral cells to correct NO-dependent endothelial dysfunction associated with hypercholesterolemia and possibly other diseases.


Assuntos
Caveolinas/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Óxido Nítrico Sintase/metabolismo , Fatores de Transcrição , Animais , Atorvastatina , Proteínas Estimuladoras de Ligação a CCAAT/antagonistas & inibidores , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Bovinos , Caveolina 1 , Células Cultivadas , Colesterol/biossíntese , LDL-Colesterol/metabolismo , LDL-Colesterol/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Endotélio Vascular/citologia , Proteínas de Choque Térmico HSP90/metabolismo , Ácidos Heptanoicos/farmacologia , Humanos , Hidroximetilglutaril-CoA Redutases/metabolismo , Leupeptinas/farmacologia , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III , Pirróis/farmacologia , Proteína de Ligação a Elemento Regulador de Esterol 1 , Regulação para Cima/efeitos dos fármacos
19.
Am J Physiol Heart Circ Physiol ; 279(3): H916-23, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10993750

RESUMO

The role of protein kinase C (PKC) isoforms in myogenic tone of the ferret coronary microcirculation was investigated by measuring fura 2 Ca(2+) signals, PKC immunoblots, contractile responses, and confocal microscopy of PKC translocation. Phorbol ester-evoked contractions were completely abolished in the absence of extracellular Ca(2+) but involved a Ca(2+) sensitization relative to KCl contractions. Immunoblotting using isoform-specific antibodies showed the presence of PKC-alpha and -iota and traces of PKC-epsilon and -mu in the ferret coronary microcirculation. PKC-beta was not detectable. When intraluminal pressure (40 to 60 and 80 mmHg) was increased, ferret coronary arterioles showed a transient increase in fura 2 Ca(2+) signals, whereas the myogenic tone remained sustained. The increase in Ca(2+) and tone was sustained at 100 mmHg. Isolated ferret coronary arterioles were fixed and immunostained for PKC-alpha at 40 and 100 mmHg intraluminal pressure. PKC translocation was determined by confocal microscopy. Increased PKC translocation was observed when vessels were exposed to 100 mmHg relative to that at resting pressure (40 mmHg). These results suggest a link between the Ca(2+) sensitization that occurs during the myogenic contraction and activation of the alpha-isoform of PKC.


Assuntos
Circulação Coronária/fisiologia , Vasos Coronários/enzimologia , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Vasoconstrição/fisiologia , Animais , Arteríolas/enzimologia , Arteríolas/fisiologia , Cálcio/metabolismo , Vasos Coronários/fisiologia , Furões , Corantes Fluorescentes , Fura-2 , Técnicas In Vitro , Masculino , Microcirculação/enzimologia , Microcirculação/fisiologia , Contração Muscular/efeitos dos fármacos , Tono Muscular/efeitos dos fármacos , Miosinas/metabolismo , Ésteres de Forbol/farmacologia , Fosforilação/efeitos dos fármacos , Cloreto de Potássio/farmacologia , Proteína Quinase C-alfa , Transdução de Sinais/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos
20.
EMBO J ; 19(16): 4272-80, 2000 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-10944110

RESUMO

In cardiac myocytes, agonist binding to muscarinic acetylcholine receptors (mAchRs) leads to the targeting of stimulated receptors to plasmalemmal microdomains termed caveolae. Here, we examined whether this translocation leads to mAchR internalization and alteration in downstream NO signaling. Differential binding of membrane-permeant and -impermeant mAchR radioligands on caveolae-enriched membranes revealed that carbachol stimulation of cardiac myocytes induces sequestration of mAchRs through caveolae fission. GTP but not its non-hydrolyzable analog GTP gamma S drove the further detachment of caveolae from myocyte sarcolemma. Also, incubation of extracts of carbachol-stimulated myocytes with recombinant GTPase dynamin induced mAchR sequestration in budded caveolae, while dominant-negative K44A dynamin inhibited it. These data were confirmed by immunofluorescence microscopy on m2 mAchR-expressing COS cells. Finally, repeated carbachol stimulations of mAchRs co-expressed in COS cells with endothelial nitric oxide synthase (eNOS) and wild-type, but not mutant, dynamin led to a progressive increase in mAchR sequestration and a concurrent stabilization of the inhibitory eNOS-caveolin complex. These findings emphasize the role of caveolae in mAchR trafficking and NO signaling, and suggest that caveolae fission may contribute to G-protein-coupled receptor desensitization.


Assuntos
Caveolinas/metabolismo , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/fisiologia , Óxido Nítrico/metabolismo , Receptores Muscarínicos/metabolismo , Transdução de Sinais , Animais , Células COS , Carbacol/farmacologia , Caveolina 1 , Membrana Celular/metabolismo , Células Cultivadas , Agonistas Colinérgicos/farmacologia , Relação Dose-Resposta a Droga , Dinaminas , Endotélio Vascular/enzimologia , Genes Dominantes , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Guanosina Trifosfato/metabolismo , Immunoblotting , Microscopia de Fluorescência , Mutagênese Sítio-Dirigida , Miocárdio/metabolismo , Óxido Nítrico Sintase/metabolismo , Ligação Proteica , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes/metabolismo , Fatores de Tempo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA