Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
JCI Insight ; 3(19)2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30282816

RESUMO

Physiological and premature aging are frequently associated with an accumulation of prelamin A, a precursor of lamin A, in the nuclear envelope of various cell types. Here, we aimed to underpin the hitherto unknown mechanisms by which prelamin A alters myonuclear organization and muscle fiber function. By experimentally studying membrane-permeabilized myofibers from various transgenic mouse lines, our results indicate that, in the presence of prelamin A, the abundance of nuclei and myosin content is markedly reduced within muscle fibers. This leads to a concept by which the remaining myonuclei are very distant from each other and are pushed to function beyond their maximum cytoplasmic capacity, ultimately inducing muscle fiber weakness.


Assuntos
Senilidade Prematura/fisiopatologia , Núcleo Celular/metabolismo , Lamina Tipo A/metabolismo , Fibras Musculares Esqueléticas/fisiologia , Força Muscular , Senilidade Prematura/genética , Animais , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Humanos , Lamina Tipo A/genética , Camundongos , Camundongos Knockout , Fibras Musculares Esqueléticas/citologia , Miosinas/metabolismo , Cultura Primária de Células
2.
Am J Physiol Lung Cell Mol Physiol ; 314(4): L670-L685, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29351439

RESUMO

Application of H2S ("sulfide") elicits a complex contraction in rat pulmonary arteries (PAs) comprising a small transient contraction (phase 1; Ph1) followed by relaxation and then a second, larger, and more sustained contraction (phase 2; Ph2). We investigated the mechanisms causing this response using isometric myography in rat second-order PAs, with Na2S as a sulfide donor. Both phases of contraction to 1,000 µM Na2S were attenuated by the pan-PKC inhibitor Gö6983 (3 µM) and by 50 µM ryanodine; the Ca2+ channel blocker nifedipine (1 µM) was without effect. Ph2 was attenuated by the mitochondrial complex III blocker myxothiazol (1 µM), the NADPH oxidase (NOX) blocker VAS2870 (10 µM), and the antioxidant TEMPOL (3 mM) but was unaffected by the complex I blocker rotenone (1 µM). The bath sulfide concentration, measured using an amperometric sensor, decreased rapidly following Na2S application, and the peak of Ph2 occurred when this had fallen to ~50 µM. Sulfide caused a transient increase in NAD(P)H autofluorescence, the offset of which coincided with development of the Ph2 contraction. Sulfide also caused a brief mitochondrial hyperpolarization (assessed using tetramethylrhodamine ethyl ester), followed immediately by depolarization and then a second more prolonged hyperpolarization, the onset of which was temporally correlated with the Ph2 contraction. Sulfide application to cultured PA smooth muscle cells increased reactive oxygen species (ROS) production (recorded using L012); this was absent when the mitochondrial flavoprotein sulfide-quinone oxoreductase (SQR) was knocked down using small interfering RNA. We propose that the Ph2 contraction is largely caused by SQR-mediated sulfide metabolism, which, by donating electrons to ubiquinone, increases electron production by complex III and thereby ROS production.


Assuntos
Benzoquinonas/química , Sulfeto de Hidrogênio/farmacologia , Músculo Liso Vascular/fisiologia , Oxirredutases/metabolismo , Artéria Pulmonar/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Sulfetos/química , Animais , Cálcio/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Músculo Liso Vascular/citologia , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Ratos , Ratos Wistar
3.
Free Radic Biol Med ; 110: 316-331, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28673614

RESUMO

The role of reactive oxygen species (ROS) in smooth muscle contraction is poorly understood. We hypothesised that G-protein coupled receptor (GPCR) activation and hypoxia induce Rho-kinase activity and contraction in rat intra-pulmonary artery (IPA) via stimulation of ROS production and subsequent Src-family kinase (SrcFK) activation. The T-type prostanoid receptor agonist U46619 induced ROS production in pulmonary artery smooth muscle cells (PASMC). U46619 also induced c-Src cysteine oxidation, SrcFK auto-phosphorylation, MYPT-1 and MLC20 phosphorylation and contraction in IPA, and all these responses were inhibited by antioxidants (ebselen, Tempol). Contraction and SrcFK/MYPT-1/MLC20 phosphorylations were also inhibited by combined superoxide dismutase and catalase, or by the SrcFK antagonist PP2, while contraction and MYPT-1/MLC20 phosphorylations were inhibited by the Rho guanine nucleotide exchange factor (RhoGEF) inhibitor Y16. H2O2 and the superoxide-generating quinoledione LY83583 both induced c-Src oxidation, SrcFK auto-phosphorylation and contraction in IPA. LY83583 and H2O2-induced contractions were inhibited by PP2, while LY83583-induced contraction was also inhibited by antioxidants and Y16. SrcFK auto-phosphorylation and MYPT-1/MLC20 phosphorylation was also induced by hypoxia in IPA and this was blocked by mitochondrial inhibitors rotenone and myxothiazol. In live PASMC, sub-cellular translocation of RhoA and the RhoGEF ARHGEF1 was triggered by both U46619 and LY83583 and this translocation was blocked by antioxidants and PP2. RhoA translocation was also inhibited by an ARHGEF1 siRNA. U46619 enhanced ROS-dependent co-immunoprecipitation of ARHGEF1 with c-Src. Our results demonstrate a link between GPCR-induced cytosolic ROS or hypoxia-induced mitochondrial ROS and SrcFK activity, Rho-kinase activity and contraction. ROS and SrcFK activate RhoA via ARHGEF1.


Assuntos
Miócitos de Músculo Liso/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Proteínas rho de Ligação ao GTP/genética , Quinases da Família src/genética , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Aminoquinolinas/farmacologia , Animais , Regulação da Expressão Gênica , Pulmão/irrigação sanguínea , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Músculo Liso/citologia , Músculo Liso/efeitos dos fármacos , Músculo Liso/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miografia , Oxirredução , Fosforilação , Cultura Primária de Células , Proteína Fosfatase 1/genética , Proteína Fosfatase 1/metabolismo , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/fisiologia , Pirimidinas/farmacologia , Ratos , Ratos Wistar , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Transdução de Sinais , Técnicas de Cultura de Tecidos , Vasoconstritores/farmacologia , Proteínas rho de Ligação ao GTP/metabolismo , Quinases da Família src/metabolismo
4.
Life Sci ; 165: 26-34, 2016 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-27686831

RESUMO

BACKGROUND AND PURPOSE: Glabridin is a major flavonoid in Glycyrrhiza glabra (licorice) root, a traditional Asian medicine. Glabridin is reported to have anti-atherogenic, anti-inflammatory and anti-nephritic properties; however its effects on vascular tone remain unexplored. EXPERIMENTAL APPROACH: We examined the effect of glabridin on rat main mesenteric artery using isometric myography and also ELISA to measure cGMP levels. KEY RESULTS: Glabridin (30µM) relaxed arteries pre-constricted with the thromboxane A2 analog U46619 (0.2µM) by ~60% in an endothelium-independent manner. Relaxation to 30µM glabridin was abolished by the guanylate cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (1µM) and by the BKCa channel blocker tetraethyammonium (1mM) but was unaffected by the estrogen receptor antagonist ICI182780. The concentration-response curve to glabridin (0.1 to 30µM) was downshifted by the KATP channel blocker glibenclamide (10µM), the KV channel blocker 4-aminopyridine (300µM), and the KIR blocker BaCl2 (30µM). In U46619-contracted arteries partially relaxed by 0.1µM sodium nitroprusside, application of 10 and 30nM glabridin caused additional vasorelaxation. Glabridin (30µM) approximately doubled tissue [cyclic GMP]. Application of the phosphodiesterase inhibitor isobutylmethylxanthine caused a much larger rise in [cyclic GMP], and glabridin failed to cause vasorelaxation or a further rise in [cGMP] when co-applied with IBMX. CONCLUSIONS AND IMPLICATIONS: Vasorelaxation to glabridin is dependent on the opening of K+ channels, particularly BKCa, probably caused by a rise in cellular [cyclic GMP] owing to phosphodiesterase inhibition. In the presence of sodium nitroprusside an effect of glabridin is observed at nM concentrations, similar those measured in plasma following human ingestion of licorice flavonoid oil.


Assuntos
AMP Cíclico/fisiologia , Isoflavonas/farmacologia , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/fisiologia , Fenóis/farmacologia , Vasodilatação/efeitos dos fármacos , Animais , Masculino , Ratos , Ratos Wistar
5.
Cardiovasc Res ; 106(1): 121-30, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25661082

RESUMO

AIMS: Sphingosylphosphorylcholine (SPC) elicits vasoconstriction at micromolar concentrations. At lower concentrations (≤1 µmol/L), however, it does not constrict intrapulmonary arteries (IPAs), but strongly potentiates vasoreactivity. Our aim was to determine whether this also occurs in a systemic artery and to delineate the signalling pathway. METHODS AND RESULTS: Rat mesenteric arteries and IPAs mounted on a myograph were challenged with ∼25 mmol/L [K+] to induce a small vasoconstriction. SPC (1 µmol/L) dramatically potentiated this constriction in all arteries by ∼400%. The potentiation was greatly suppressed or abolished by inhibition of phospholipase C (PLC; U73122), PKCε (inhibitory peptide), Src (PP2), and NADPH oxidase (VAS2870), and also by Tempol (superoxide scavenger), but not by inhibition of Rho kinase (Y27632). Potentiation was lost in mesenteric arteries from p47(phox-/-), but not NOX2(-/-), mice. The intracellular superoxide generator LY83583 mimicked the effect of SPC. SPC elevated reactive oxygen species (ROS) in vascular smooth muscle cells, and this was blocked by PP2, VAS2870, and siRNA knockdown of PKCε. SPC (1 µmol/L) significantly reduced the EC50 for U46619-induced vasoconstriction, an action ablated by Tempol. In patch-clamped mesenteric artery cells, SPC (200 nmol/L) enhanced Ba2+ current through L-type Ca2+ channels, an action abolished by Tempol but mimicked by LY83583. CONCLUSION: Our results suggest that low concentrations of SPC activate a PLC-coupled and NOX1-mediated increase in ROS, with consequent enhancement of voltage-gated Ca2+ entry and thus vasoreactivity. We speculate that this pathway is not specific for SPC, but may also contribute to vasoconstriction elicited by other G-protein coupled receptor and PLC-coupled agonists.


Assuntos
Canais de Cálcio/efeitos dos fármacos , Artérias Mesentéricas/fisiologia , NADH NADPH Oxirredutases/fisiologia , Fosforilcolina/análogos & derivados , Artéria Pulmonar/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Esfingosina/análogos & derivados , Vasoconstrição/efeitos dos fármacos , Animais , Canais de Cálcio/fisiologia , Óxidos N-Cíclicos/farmacologia , Relação Dose-Resposta a Droga , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Artérias Mesentéricas/efeitos dos fármacos , Camundongos , Camundongos Knockout , Modelos Animais , NADPH Oxidase 1 , NADPH Oxidase 2 , NADPH Oxidases/deficiência , NADPH Oxidases/genética , NADPH Oxidases/farmacologia , NADPH Oxidases/fisiologia , Fosforilcolina/farmacologia , Proteína Quinase C-épsilon/farmacologia , Artéria Pulmonar/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Esfingosina/farmacologia , Marcadores de Spin , Fosfolipases Tipo C/farmacologia , Vasoconstrição/fisiologia
6.
Cardiovasc Res ; 99(3): 404-11, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23708740

RESUMO

AIMS: To determine the role of gap junctions (GJs) in hypoxic pulmonary vasoconstriction (HPV). METHODS AND RESULTS: Studies were performed in rat isolated intrapulmonary arteries (IPAs) mounted on a myograph and in anaesthetized rats. Hypoxia induced a biphasic HPV response in IPAs preconstricted with prostaglandin F2α (PGF2α, 3 µM) or 20 mM K⁺. The GJ inhibitors 18ß-glycyrrhetinic acid (18ß-GA, 30 µM), heptanol (3.5 mM), or 2-aminoethoxydiphenyl borate (2-APB) (75 µM) had little effect on the transient Phase 1 of HPV, but abolished the sustained Phase 2 which is associated with Ca²âº sensitization. The voltage-dependent Ca²âº channel blocker diltiazem (10 µM) had no effect on HPV, and did not alter the inhibitory action of 18ß-GA. Sustained HPV is enhanced by high glucose (15 mM) via potentiation of Ca²âº sensitization, in the presence of high glucose 18ß-GA still abolished sustained HPV. Simultaneous measurement of tension and intracellular Ca²âº using Fura PE-3 demonstrated that whilst 18ß-GA abolished tension development during sustained HPV, it did not affect the elevation of intracellular Ca²âº. Consistent with this, 18ß-GA abolished hypoxia-induced phosphorylation of the Rho kinase target MYPT-1. In anaesthetized rats hypoxia caused a biphasic increase in systolic right ventricular pressure. Treatment with oral 18ß-GA (25 mg/kg) abolished the sustained component of the hypoxic pressor response. CONCLUSION: These results imply that GJs are critically involved in the signalling pathways leading to Rho kinase-dependent Ca²âº sensitization during sustained HPV, but not elevation of intracellular Ca²âº, and may explain the dependence of the former on an intact endothelium.


Assuntos
Cálcio/metabolismo , Junções Comunicantes/metabolismo , Hipóxia/fisiopatologia , Artéria Pulmonar/fisiopatologia , Vasoconstrição/fisiologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/metabolismo , Diltiazem/farmacologia , Junções Comunicantes/efeitos dos fármacos , Ácido Glicirretínico/análogos & derivados , Ácido Glicirretínico/farmacologia , Hipóxia/metabolismo , Masculino , Fosforilação , Proteína Fosfatase 1/metabolismo , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Quinases Associadas a rho/metabolismo
7.
Cardiovasc Res ; 89(1): 214-24, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20805095

RESUMO

AIMS: the aim of this study was to determine the relative importance of Ca(2+) sensitization, ion channels, and intracellular Ca(2+) ([Ca(2+)](i)) in the mixed constrictor/relaxation actions of superoxide anion on systemic and pulmonary arteries. METHODS AND RESULTS: pulmonary and mesenteric arteries were obtained from rat. Superoxide was generated in arteries and cells with 6-anilino-5,8-quinolinequinone (LY83583). Following pre-constriction with U46619, 10 µmol/L LY83583 caused constriction in pulmonary and relaxation in mesenteric arteries. Both constrictor and relaxant actions of LY83583 were inhibited by superoxide dismutase and catalase. LY83583 caused Rho-kinase-dependent constriction in α-toxin-permeabilized pulmonary but not mesenteric arteries. Phosphorylation of myosin phosphatase-targeting subunit-1 (MYPT-1; as determined by western blot), was enhanced by LY83583 in pulmonary artery only. However, in both artery types, changes in tension were closely correlated with changes in phosphorylation of the 20 kDa myosin light chain as well as changes in [Ca(2+)](i) (as measured with Fura PE-3), with LY83583 causing increases in pulmonary and decreases in mesenteric arteries. When U46619 was replaced by 30 mmol/L K(+), all changes in [Ca(2+)](i) were abolished and LY83583 constricted both artery types. The K(V) channel inhibitor 4-aminopyridine abolished the LY83583-induced relaxation in mesenteric artery without affecting constriction in pulmonary artery. However, LY83583 caused a similar hyperpolarizing shift in the steady-state activation of K(V) current in isolated smooth muscle cells of both artery types. CONCLUSIONS: superoxide only causes Rho-kinase-dependent Ca(2+) sensitization in pulmonary artery, resulting in constriction, and whilst it opens K(V) channels in both artery types, this only results in relaxation in mesenteric.


Assuntos
Artérias Mesentéricas/fisiologia , Artéria Pulmonar/fisiologia , Superóxidos/metabolismo , Vasoconstrição/fisiologia , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , 4-Aminopiridina/farmacologia , Aminoquinolinas/farmacologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Guanilato Ciclase/antagonistas & inibidores , Técnicas In Vitro , Masculino , Artérias Mesentéricas/citologia , Artérias Mesentéricas/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Oxidiazóis/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Quinoxalinas/farmacologia , Ratos , Ratos Wistar , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Quinases Associadas a rho/metabolismo
8.
Stem Cells ; 28(11): 1950-60, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20872847

RESUMO

Neural induction is the first step in the formation of the vertebrate central nervous system. The emerging consensus of the mechanisms underlying neural induction is the combined influences from inhibiting bone morphogenetic protein (BMP) signaling and activating fibroblast growth factor (FGF)/Erk signaling, which act extrinsically via either autocrine or paracrine fashions. However, do intrinsic forces (cues) exist and do they play decisive roles in neural induction? These questions remain to be answered. Here, we have identified a novel neural initiator, neuronatin (Nnat), which acts as an intrinsic factor to promote neural fate in mammals and Xenopus. ESCs lacking this intrinsic factor fail to undergo neural induction despite the inhibition of the BMP pathway. We show that Nnat initiates neural induction in ESCs through increasing intracellular Ca(2+) ([Ca(2+) ](i)) by antagonizing Ca(2+) -ATPase isoform 2 (sarco/endoplasmic reticulum Ca(2+) -ATPase isoform 2) in the endoplasmic reticulum, which in turn increases the phosphorylation of Erk1/2 and inhibits the BMP4 pathway and leads to neural induction in conjunction with FGF/Erk pathway.


Assuntos
Sinalização do Cálcio/fisiologia , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Western Blotting , Sinalização do Cálcio/genética , Diferenciação Celular/genética , Citometria de Fluxo , Imuno-Histoquímica , Imunoprecipitação , Proteínas de Membrana/genética , Camundongos , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Neurônios/metabolismo , Fosforilação , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo
9.
J Neurochem ; 114(3): 832-42, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20477946

RESUMO

Epidemiological studies have shown an association between statin use and a decreased risk of dementia. However, the mechanism by which this beneficial effect is brought about is unclear. In the context of Alzheimer's disease, at least three possibilities have been studied; reduction in amyloid beta peptide (Abeta) production, the promotion of alpha-secretase cleavage and positive effects on neurite outgrowth. By investigating the effects of mevalonate pathway blockade on neurite outgrowth using real-time imaging, we found that rather than promote the production of neurite extensions, inhibition rapidly induced cell rounding. Crucially, neurite-like structures were generated through the persistence of cell-cell and cell-substrate adhesions and not through a mechanism of positive outgrowth. This effect can be strikingly enhanced by the over-expression of human amyloid precursor protein and is isoprenoid rather than cholesterol dependent.


Assuntos
Precursor de Proteína beta-Amiloide/fisiologia , Ácido Mevalônico/antagonistas & inibidores , Neuritos/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Forma Celular/fisiologia , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Ácido Mevalônico/metabolismo , Microscopia de Vídeo/métodos , Neuritos/efeitos dos fármacos , Neuritos/patologia , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia
10.
Proc Natl Acad Sci U S A ; 106(26): 10775-80, 2009 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-19541629

RESUMO

Phenotypic modulation of airway smooth muscle (ASM) is an important feature of airway remodeling in asthma that is characterized by enhanced proliferation and secretion of pro-inflammatory chemokines. These activities are regulated by the concentration of free Ca(2+) in the cytosol ([Ca(2+)](i)). A rise in [Ca(2+)](i) is normalized by rapid reuptake of Ca(2+) into sarcoplasmic reticulum (SR) stores by the sarco/endoplasmic reticulum Ca(2+) (SERCA) pump. We examined whether increased proliferative and secretory responses of ASM from asthmatics result from reduced SERCA expression. ASM cells were cultured from subjects with and without asthma. SERCA expression was evaluated by western blot, immunohistochemistry and real-time PCR. Changes in [Ca(2+)](i), cell spreading, cellular proliferation, and eotaxin-1 release were measured. Compared with control cells from healthy subjects, SERCA2 mRNA and protein expression was reduced in ASM cells from subjects with moderately severe asthma. SERCA2 expression was similarly reduced in ASM in vivo in subjects with moderate/severe asthma. Rises in [Ca(2+)](i) following cell surface receptor-induced SR activation, or inhibition of SERCA-mediated Ca(2+) re-uptake, were attenuated in ASM cells from asthmatics. Likewise, the return to baseline of [Ca](i) after stimulation by bradykinin was delayed by approximately 50% in ASM cells from asthmatics. siRNA-mediated knockdown of SERCA2 in ASM from healthy subjects increased cell spreading, eotaxin-1 release and proliferation. Our findings implicate a deficiency in SERCA2 in ASM in asthma that contributes to its secretory and hyperproliferative phenotype in asthma, and which may play a key role in mechanisms of airway remodeling.


Assuntos
Asma/metabolismo , Brônquios/metabolismo , Retículo Sarcoplasmático/enzimologia , Asma/patologia , Asma/fisiopatologia , Western Blotting , Brônquios/patologia , Brônquios/fisiopatologia , Cálcio/metabolismo , Movimento Celular , Proliferação de Células , Células Cultivadas , Quimiocina CCL11/metabolismo , Feminino , Regulação Enzimológica da Expressão Gênica , Homeostase , Humanos , Imuno-Histoquímica , Interleucina-13/farmacologia , Masculino , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático
11.
Free Radic Biol Med ; 46(5): 633-42, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19103285

RESUMO

Reactive oxygen species play a key role in vascular disease, pulmonary hypertension, and hypoxic pulmonary vasoconstriction. We investigated contractile responses, intracellular Ca(2+) ([Ca(2+)](i)), Rho-kinase translocation, and phosphorylation of the regulatory subunit of myosin phosphatase (MYPT-1) and of myosin light chain (MLC(20)) in response to LY83583, a generator of superoxide anion, in small intrapulmonary arteries (IPA) of rat. LY83583 caused concentration-dependent constrictions in IPA and greatly enhanced submaximal PGF(2alpha)-mediated preconstriction. In small femoral or mesenteric arteries of rat, LY83583 alone was without effect, but it relaxed a PGF(2)alpha-mediated preconstriction. Constrictions in IPA were inhibited by superoxide dismutase and tempol, but not catalase, and were endothelium and guanylate cyclase independent. Constrictions were also inhibited by the Rho-kinase inhibitor Y27632 and the Src-family kinase inhibitor SU6656. LY83583 did not raise [Ca(2+)](i), but caused a Y27632-sensitive constriction in alpha-toxin-permeabilized IPA. LY83583 triggered translocation of Rho-kinase from the nucleus to the cytosol in pulmonary artery smooth muscle cells and enhanced phosphorylation of MYPT-1 at Thr-855 and of MLC(20) at Ser-19 in IPA. This enhancement was inhibited by superoxide dismutase and abolished by Y27632. Hydrogen peroxide did not activate Rho-kinase. We conclude that in rat small pulmonary artery, superoxide triggers Rho-kinase-mediated Ca(2+) sensitization and vasoconstriction independent of hydrogen peroxide.


Assuntos
Broncoconstrição/fisiologia , Hipertensão Pulmonar/enzimologia , Miócitos de Músculo Liso/fisiologia , Superóxidos/farmacologia , Quinases Associadas a rho/fisiologia , Quinases da Família src/fisiologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Amidas/farmacologia , Aminoquinolinas/farmacologia , Animais , Broncoconstrição/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Guanilato Ciclase/antagonistas & inibidores , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/prevenção & controle , Indóis/farmacologia , Masculino , Miócitos de Músculo Liso/patologia , Cadeias Leves de Miosina/metabolismo , Fosforilação , Proteína Fosfatase 1/metabolismo , Artéria Pulmonar/patologia , Piridinas/farmacologia , Ratos , Ratos Wistar , Sulfonamidas/farmacologia , Quinases Associadas a rho/antagonistas & inibidores , Quinases da Família src/antagonistas & inibidores
12.
Free Radic Biol Med ; 45(10): 1468-76, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18805479

RESUMO

Reactive oxygen species are implicated in pulmonary hypertension and hypoxic pulmonary vasoconstriction. We examined the effects of low concentrations of peroxide on intrapulmonary arteries (IPA). IPAs from Wistar rats were mounted on a myograph for recording tension and estimating intracellular Ca2+ using Fura-PE3. Ca2+ sensitization was examined in alpha-toxin-permeabilized IPAs, and phosphorylation of MYPT-1 and MLC(20) was assayed by Western blot. Peroxide (30 microM) induced a vasoconstriction with transient and sustained components and equivalent elevations of intracellular Ca2+. The transient constriction was strongly suppressed by indomethacin, the TP-receptor antagonist SQ-29584, and the Rho kinase inhibitor Y-27632, whereas sustained constriction was unaffected. Neither vasoconstriction nor elevation of intracellular Ca2+ was affected by removal of extracellular Ca2+, whereas dantrolene suppressed the former and ryanodine abolished the latter. Peroxide-induced constriction of permeabilized IPAs was unaffected by Y-27632 but abolished by PKC inhibitors; these also suppressed constriction in intact IPAs. Peroxide caused translocation of PKCalpha, but had no significant effect on MYPT-1 or MLC(20) phosphorylation. We conclude that in IPAs peroxide causes transient release of vasoconstrictor prostanoids, but sustained constriction is associated with release of Ca2+ from ryanodine-sensitive stores and a PKC-dependent but Rho kinase- and MLC(20)-independent constrictor mechanism.


Assuntos
Cálcio/metabolismo , Peróxido de Hidrogênio/farmacologia , Proteína Quinase C/metabolismo , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/fisiologia , Vasoconstrição/efeitos dos fármacos , Animais , Masculino , Ratos , Ratos Wistar , Fatores de Tempo
13.
Cardiovasc Res ; 80(3): 453-62, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18682436

RESUMO

AIMS: We investigated the role of src-family kinases (srcFKs) in hypoxic pulmonary vasoconstriction (HPV) and how this relates to Rho-kinase-mediated Ca(2+) sensitization and changes in intracellular Ca(2+) concentration ([Ca(2+)](i)). METHODS AND RESULTS: Intra-pulmonary arteries (IPAs) were obtained from male Wistar rats. HPV was induced in myograph-mounted IPAs. Auto-phosphorylation of srcFKs and phosphorylation of the regulatory subunit of myosin phosphatase (MYPT-1) and myosin light-chain (MLC(20)) in response to hypoxia were determined by western blotting. Translocation of Rho-kinase and effects of siRNA knockdown of src and fyn were examined in cultured pulmonary artery smooth muscle cells (PASMCs). [Ca(2+)](i) was estimated in Fura-PE3-loaded IPA. HPV was inhibited by two blockers of srcFKs, SU6656 and PP2. Hypoxia enhanced phosphorylation of three srcFK proteins at Tyr-416 (60, 59, and 54 kDa, corresponding to src, fyn, and yes, respectively) and enhanced srcFK-dependent tyrosine phosphorylation of multiple target proteins. Hypoxia caused a complex, time-dependent enhancement of MYPT-1 and MLC(20) phosphorylation, both in the absence and presence of pre-constriction. The sustained component of this enhancement was blocked by SU6656 and the Rho-kinase inhibitor Y27632. In PASMCs, hypoxia caused translocation of Rho-kinase from the nucleus to the cytoplasm, and this was prevented by anti-src siRNA and to a lesser extent by anti-fyn siRNA. The biphasic increases in [Ca(2+)](i) that accompany HPV were also inhibited by PP2. CONCLUSION: Hypoxia activates srcFKs and triggers protein tyrosine phosphorylation in IPA. Hypoxia-mediated Rho-kinase activation, Ca(2+) sensitization, and [Ca(2+)](i) responses are depressed by srcFK inhibitors and/or siRNA knockdown, suggesting a central role of srcFKs in HPV.


Assuntos
Hipóxia/metabolismo , Artéria Pulmonar/metabolismo , Vasoconstrição/fisiologia , Quinases da Família src/metabolismo , Amidas/farmacologia , Animais , Cálcio/metabolismo , Inibidores Enzimáticos/farmacologia , Indóis/farmacologia , Masculino , Modelos Animais , Cadeias Leves de Miosina/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Fosforilação , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Piridinas/farmacologia , Ratos , Ratos Wistar , Sulfonamidas/farmacologia , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo , Quinases da Família src/antagonistas & inibidores
14.
Cardiovasc Res ; 77(3): 570-9, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18032393

RESUMO

AIMS: We investigated the role of src family kinases (srcFK) in agonist-mediated Ca2+-sensitization in pulmonary artery and whether this involves interaction with the rho/rho-kinase pathway. METHODS AND RESULTS: Intra-pulmonary arteries (IPAs) and cultured pulmonary artery smooth muscle cells (PASMC) were obtained from rat. Expression of srcFK was determined at the mRNA and protein levels. Ca2+-sensitization was induced by prostaglandin F(2 alpha) (PGF(2 alpha)) in alpha-toxin-permeabilized IPAs. Phosphorylation of the regulatory subunit of myosin phosphatase (MYPT-1) and of myosin light-chain-20 (MLC20) and translocation of rho-kinase in response to PGF(2 alpha) were also determined. Nine srcFK were expressed at the mRNA level, including src, fyn, and yes, and PGF(2 alpha) enhanced phosphorylation of three srcFK proteins at tyr-416. In alpha-toxin-permeabilized IPAs, PGF(2 alpha) enhanced the Ca2+-induced contraction (pCa 6.9) approximately three-fold. This enhancement was inhibited by the srcFK blockers SU6656 and PP2 and by the rho-kinase inhibitor Y27632. Y27632, but not SU6656 or PP2, also inhibited the underlying pCa 6.9 contraction. PGF(2 alpha) enhanced phosphorylation of MYPT-1 at thr-697 and thr-855 and of MLC20 at ser-19. This enhancement, but not the underlying basal phosphorylation, was inhibited by SU6656. Y27632 suppressed both basal and PGF(2 alpha)-mediated phosphorylation. The effects of SU6656 and Y27632, on both contraction and MYPT-1 and MLC20 phosphorylation, were not additive. PGF(2 alpha) triggered translocation of rho-kinase in PASMC, and this was inhibited by SU6656. CONCLUSIONS: srcFK are activated by PGF(2 alpha) in the rat pulmonary artery and may contribute to Ca2+-sensitization and contraction via rho-kinase translocation and phosphorylation of MYPT-1.


Assuntos
Cálcio/metabolismo , Artéria Pulmonar/metabolismo , Quinases Associadas a rho/fisiologia , Quinases da Família src/fisiologia , Animais , Células Cultivadas , Dinoprosta/farmacologia , Masculino , Cadeias Leves de Miosina/metabolismo , Fosforilação , Proteína Fosfatase 1/metabolismo , Ratos , Ratos Wistar , Tirosina/metabolismo
15.
Hypertension ; 51(2): 239-45, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18158336

RESUMO

Sphingosylphosphorylcholine (SPC) is a powerful vasoconstrictor, but in vitro its EC(50) is approximately 100-fold more than plasma concentrations. We examined whether subcontractile concentrations of SPC (100 nmol/L of SPC, and independent of the endothelium, 2-aminoethoxydiphenylborane-sensitive Ca(2+) entry, and Rho kinase. It was abolished by the phospholipase C inhibitor U73122, the broad spectrum protein kinase C (PKC) inhibitor Ro31-8220, and the PKC delta inhibitor rottlerin, but not by Gö6976, which is ineffective against PKC delta. The potentiation could be attributed to enhancement of Ca(2+) entry. SPC also potentiated the responses to prostaglandin F(2 alpha) and U436619, which activate a 2-aminoethoxydiphenylborane sensitive nonselective cation channel in intrapulmonary arteries. In this case, potentiation was partially inhibited by diltiazem but abolished by 2-aminoethoxydiphenylborane, Ro31-8220, and rottlerin. SPC (1 micromol/L) caused translocation of PKC delta to the perinuclear region and cytoskeleton of cultured intrapulmonary artery smooth muscle cells. We present the novel finding that low, subcontractile concentrations of SPC potentiate Ca(2+) entry in intrapulmonary arteries through both voltage-dependent and independent pathways via a receptor-dependent mechanism involving PKC delta. This has implications for the physiological role of SPC, especially in cardiovascular disease, where SPC is reported to be elevated.


Assuntos
Cálcio/metabolismo , Fosforilcolina/análogos & derivados , Proteína Quinase C-delta/fisiologia , Artéria Pulmonar/fisiologia , Esfingosina/análogos & derivados , Sistema Vasomotor/efeitos dos fármacos , Animais , Transporte Biológico/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Células Cultivadas , Diltiazem/farmacologia , Dinoprosta/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Eletrofisiologia , Inibidores Enzimáticos/farmacologia , Membranas Intracelulares/metabolismo , Masculino , Miócitos de Músculo Liso/enzimologia , Concentração Osmolar , Fosforilcolina/administração & dosagem , Fosforilcolina/farmacologia , Potássio/farmacologia , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/metabolismo , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/enzimologia , Artéria Pulmonar/metabolismo , Ratos , Ratos Wistar , Esfingosina/administração & dosagem , Esfingosina/farmacologia , Vasoconstrição/efeitos dos fármacos
16.
Am J Physiol Lung Cell Mol Physiol ; 293(4): L982-90, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17616643

RESUMO

It has been proposed that a hypoxia-induced inhibition of the Na(+)-Ca(2+) exchanger (NCX) contributes to hypoxic pulmonary vasoconstriction (HPV). By recording isometric tension development in rat intrapulmonary arteries (IPA), we examined the effect on HPV of maneuvers that reduce the ability of NCX to regulate intracellular Ca(2+) concentration ([Ca(2+)](i)). In some experiments, fura pentakis(acetoxymethyl) ester-3 (fura PE-3) was also used to monitor [Ca(2+)](i). HPV was elicited in IPA that were pretreated with 10 microM diltiazem and slightly preconstricted with PGF(2alpha), which enhances the hypoxic response. Substitution of Na(+) with Li(+) increased HPV and the associated rise in [Ca(2+)](i). Pretreatment with ouabain (100 microM) to diminish the Na(+) gradient or with the reverse-mode NCX inhibitor KB-R7943 (3 or 10 microM) had no significant effect on HPV. Combined treatment with ouabain and low-[Na(+)] (24 mM) solution enhanced HPV strongly. The role of NCX in Ca(2+) extrusion was examined by assessing the decrease in [Ca(2+)](i) in Ca(2+)-free physiological saline solution either containing or lacking Na(+) following a high K(+)-induced loading of cellular [Ca(2+)]. Although the large initial rapid fall in [Ca(2+)] was Na(+) independent, final recovery of [Ca(2+)] to its basal level was delayed in the absence of Na(+). Therefore, HPV persisted or was increased under conditions in which forward-mode NCX was already attenuated or prevented, demonstrating that inhibition of NCX by hypoxia is unlikely to initiate HPV. Instead, NCX appears to act to inhibit HPV as would be expected if it is functioning to extrude Ca(2+).


Assuntos
Hipóxia/fisiopatologia , Artéria Pulmonar/fisiopatologia , Trocador de Sódio e Cálcio/metabolismo , Vasoconstrição , Animais , Cálcio/metabolismo , Membranas Intracelulares/metabolismo , Contração Isométrica , Lítio/farmacologia , Masculino , Concentração Osmolar , Potássio/farmacologia , Artéria Pulmonar/metabolismo , Ratos , Ratos Wistar , Sódio/farmacologia , Vasoconstrição/efeitos dos fármacos
17.
Novartis Found Symp ; 272: 259-68; discussion 268-79, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16686440

RESUMO

The identities of the Ca2+ influx mechanisms underlying hypoxic pulmonary vasoconstriction (HPV), the contractile response of pulmonary arteries (PA) to hypoxia, remain controversial. We investigated the roles of Na+/Ca2+ exchange (NCX) and capacitative Ca2+ entry (CCE) in HPV by measuring isometric tension in rat intrapulmonary arteries (IPA) during hypoxia. It has been shown in PA cells that hypoxia raised [Ca2+]i by inhibiting NCX. We found that removal of Na+ caused a response resembling HPV, suggesting that HPV could be due to inhibition of NCX. However, prior inhibition of NCX using Na+ free solution or 3 microM KB-R7943 enhanced rather than prevented HPV, indicating that HPV is not caused by an inhibition of NCX. The CCE blocker 2-APB inhibited both phases of HPV, and the ryanodine receptor blocker dantrolene inhibited the 2nd phase of HPV. Taken together with previous observations, these results suggest a role for CCE in HPV, but suggest that different CCE pathways may be involved for each phase.


Assuntos
Hipóxia/metabolismo , Pulmão/metabolismo , Pulmão/fisiopatologia , Artéria Pulmonar , Trocador de Sódio e Cálcio , Vasoconstrição , Animais , Pulmão/irrigação sanguínea , Artéria Pulmonar/metabolismo , Artéria Pulmonar/fisiopatologia , Ratos , Trocador de Sódio e Cálcio/metabolismo , Trocador de Sódio e Cálcio/fisiologia , Vasoconstrição/fisiologia
18.
J Physiol ; 570(Pt 1): 53-8, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16254010

RESUMO

The pulmonary circulation differs from the systemic in several important aspects, the most important being that pulmonary arteries constrict to moderate physiological (20-60 mmHg PO2) hypoxia, whereas systemic arteries vasodilate. This phenomenon is called hypoxic pulmonary vasoconstriction (HPV), and is responsible for maintaining the ventilation-perfusion ratio during localized alveolar hypoxia. In disease, however, global hypoxia results in a detrimental increase in total pulmonary vascular resistance, and increased load on the right heart. Despite many years of study, the precise mechanisms underlying HPV remain unresolved. However, as we argue below, there is now overwhelming evidence that hypoxia can stimulate several pathways leading to a rise in the intracellular Ca2+ concentration ([Ca2+]i) in pulmonary artery smooth muscle cells (PASMC). This rise in [Ca2+]i is consistently found to be relatively small, and HPV seems also to require rho kinase-mediated Ca2+ sensitization. There is good evidence that HPV also has an as yet unexplained endothelium dependency. In this brief review, we highlight selected recent findings and ongoing controversies which continue to animate the study of this remarkable and unique response of the pulmonary vasculature to hypoxia.


Assuntos
Hipóxia Celular/fisiologia , Circulação Pulmonar/fisiologia , Vasoconstrição/fisiologia , Animais , Cálcio/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Humanos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo
19.
J Physiol ; 571(Pt 1): 147-63, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16357015

RESUMO

The mechanisms by which prostaglandin F(2alpha) (PGF(2alpha)) increases intracellular Ca2+ concentration [Ca2+]i in vascular smooth muscle remain unclear. We examined the role of store-, receptor- and voltage-operated Ca2+ influx pathways in rat intrapulmonary arteries (IPA) loaded with Fura PE-3. Low concentrations (0.01-1 microM) of PGF(2alpha) caused a transient followed by a plateau rise in [Ca2+]i. Both responses became maximal at 0.1 microM PGF(2alpha). At higher concentrations of PGF(2alpha), a further slower rise in [Ca2+]i was superimposed on the plateau. The [Ca2+]i response to 0.1 microM PGF(2alpha) was mimicked by the FP receptor agonist fluprostenol, whilst the effect of 10 microM PGF(2alpha) was mimicked by the TP receptor agonist U-46619. The plateau rise in [Ca2+]i in response to 0.1 microM PGF(2alpha) was insensitive to diltiazem, and was abolished in Ca2+-free physiological salt solution, and by pretreatment with La3+, 2-APB, thapsigargin or U-73122. The rises in [Ca2+]i in response to 10 microM PGF(2alpha) and 0.01 microM U-46619 were partially inhibited by diltiazem. The diltiazem-resistant components of both of these responses were inhibited by 2-APB and La3+ to an extent which was significantly less than that seen for the response to 0.1 microM PGF(2alpha), and were also much less sensitive to U-73122. The U-46619 response was also relatively insensitive to thapsigargin. When Ca2+ was replaced with Sr2+, the sustained increase in the Fura PE-3 signal to 0.1 microM PGF(2alpha) was abolished, whereas 10 microM PGF(2alpha) and 0.05 microM U-46619 still caused substantial increases. These results suggest that low concentrations of PGF(2alpha) act via FP receptors to cause IP3-dependent Ca2+ release and store operated Ca2+ entry (SOCE). U-46619 and 10-100 microM PGF(2alpha) cause a TP receptor-mediated Ca2+ influx involving both L-type Ca2+ channels and a receptor operated pathway, which differs from SOCE in its susceptibility to La3+, 2-APB and thapsigargin, does not require phospholipase C activation, and is Sr2+ permeable.


Assuntos
Cálcio/análise , Cálcio/metabolismo , Dinoprosta/farmacologia , Artéria Pulmonar/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Animais , Compostos de Boro/farmacologia , Canais de Cálcio Tipo L/fisiologia , Fármacos Cardiovasculares/farmacologia , Diltiazem/farmacologia , Inositol 1,4,5-Trifosfato/fisiologia , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Artéria Pulmonar/química , Ratos , Ratos Wistar , Receptores de Prostaglandina/efeitos dos fármacos , Receptores de Prostaglandina/fisiologia , Receptores de Tromboxanos/efeitos dos fármacos , Receptores de Tromboxanos/fisiologia , Transdução de Sinais/fisiologia , Fosfolipases Tipo C/farmacologia , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia
20.
Am J Physiol Lung Cell Mol Physiol ; 289(6): L1039-48, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16055481

RESUMO

The mechanisms through which p38 mitogen-activated protein kinase (p38 MAPK) is involved in smooth muscle contraction remain largely unresolved. We examined the role of p38 MAPK in prostaglandin F(2alpha) (PGF(2alpha))-induced vasoconstriction and in hypoxic pulmonary vasoconstriction (HPV) of rat small intrapulmonary arteries (IPA). The p38 MAPK inhibitors SB-203580 and SB-202190 strongly inhibited PGF(2alpha)-induced vasoconstriction, with IC(50)s of 1.6 and 1.2 microM, whereas the inactive analog SB-202474 was approximately 30-fold less potent. Both transient and sustained phases of HPV were suppressed by SB-203580, but not by SB-202474 (both 2 microM). Western blot analysis revealed that PGF(2alpha) (20 microM) increased phosphorylation of p38 MAPK and of heat shock protein 27 (HSP27), and this was abolished by SB-203580 but not by SB-202474 (both 2 microM). Endothelial denudation or blockade of endothelial nitric oxide (NO) synthase with N(omega)-nitro-L-arginine methyl ester (L-NAME) significantly suppressed the relaxation of PGF(2alpha)-constricted IPA by SB-203580, but not by SB-202474. Similarly, the inhibition of HPV by SB-203580 was prevented by prior treatment with L-NAME. SB-203580 (2 microM), but not SB-202474, enhanced relaxation-induced by the NO donor S-nitroso-N-acetylpenicillamine (SNAP) in endothelium-denuded IPA constricted with PGF(2alpha). In alpha-toxin-permeabilized IPA, SB-203580-induced relaxation occurred in the presence but not the absence of the NO donor sodium nitroprusside (SNP); SB-202474 was without effect even in the presence of SNP. In intact IPA, neither PGF(2alpha)- nor SNAP-mediated changes in cytosolic free Ca(2+) were affected by SB-203580. We conclude that p38 MAPK contributes to PGF(2alpha)- and hypoxia-induced constriction of rat IPA primarily by antagonizing the underlying Ca(2+)-desensitizing actions of NO.


Assuntos
Dinoprosta/farmacologia , Hipóxia/metabolismo , Óxido Nítrico/metabolismo , Ocitócicos/administração & dosagem , Artéria Pulmonar/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Constrição Patológica/induzido quimicamente , Constrição Patológica/metabolismo , Inibidores Enzimáticos/farmacologia , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/metabolismo , Masculino , Contração Muscular/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/metabolismo , Técnicas de Cultura de Órgãos/métodos , Fosforilação/efeitos dos fármacos , Ratos , Ratos Wistar , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA