Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur Respir J ; 60(2)2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35058244

RESUMO

In this review article we present the evidence to date supporting the role of the calcium-sensing receptor (CaSR) as a key, pluripotential molecular trigger for asthma and speculate on the likely benefits of topical therapy of asthma with negative allosteric modulators of the CaSR: calcilytics.


Assuntos
Asma , Receptores de Detecção de Cálcio , Asma/tratamento farmacológico , Cálcio , Humanos
2.
J Pharmacol Exp Ther ; 376(1): 51-63, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33115824

RESUMO

Asthma is still an incurable disease, and there is a recognized need for novel small-molecule therapies for people with asthma, especially those poorly controlled by current treatments. We previously demonstrated that calcium-sensing receptor (CaSR) negative allosteric modulators (NAMs), calcilytics, uniquely suppress both airway hyperresponsiveness (AHR) and inflammation in human cells and murine asthma surrogates. Here we assess the feasibility of repurposing four CaSR NAMs, which were originally developed for oral therapy for osteoporosis and previously tested in the clinic as a novel, single, and comprehensive topical antiasthma therapy. We address the hypotheses, using murine asthma surrogates, that topically delivered CaSR NAMs 1) abolish AHR; 2) are unlikely to cause unwanted systemic effects; 3) are suitable for topical application; and 4) inhibit airway inflammation to the same degree as the current standard of care, inhaled corticosteroids, and, furthermore, inhibit airway remodeling. All four CaSR NAMs inhibited poly-L-arginine-induced AHR in naïve mice and suppressed both AHR and airway inflammation in a murine surrogate of acute asthma, confirming class specificity. Repeated exposure to inhaled CaSR NAMs did not alter blood pressure, heart rate, or serum calcium concentrations. Optimal candidates for repurposing were identified based on anti-AHR/inflammatory activities, pharmacokinetics/pharmacodynamics, formulation, and micronization studies. Whereas both inhaled CaSR NAMs and inhaled corticosteroids reduced airways inflammation, only the former prevented goblet cell hyperplasia in a chronic asthma model. We conclude that inhaled CaSR NAMs are likely a single, safe, and effective topical therapy for human asthma, abolishing AHR, suppressing airways inflammation, and abrogating some features of airway remodeling. SIGNIFICANCE STATEMENT: Calcium-sensing receptor (CaSR) negative allosteric modulators (NAMs) reduce airway smooth muscle hyperresponsiveness, reverse airway inflammation as efficiently as topical corticosteroids, and suppress airway remodeling in asthma surrogates. CaSR NAMs, which were initially developed for oral therapy of osteoporosis proved inefficacious for this indication despite being safe and well tolerated. Here we show that structurally unrelated CaSR NAMs are suitable for inhaled delivery and represent a one-stop, steroid-free approach to asthma control and prophylaxis.


Assuntos
Antiasmáticos/uso terapêutico , Asma/tratamento farmacológico , Indanos/uso terapêutico , Naftalenos/uso terapêutico , Fenilpropionatos/uso terapêutico , Quinazolinonas/uso terapêutico , Receptores de Detecção de Cálcio/agonistas , Regulação Alostérica , Animais , Antiasmáticos/efeitos adversos , Antiasmáticos/farmacologia , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Reposicionamento de Medicamentos , Células HEK293 , Humanos , Indanos/efeitos adversos , Indanos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Naftalenos/efeitos adversos , Naftalenos/farmacologia , Fenilpropionatos/efeitos adversos , Fenilpropionatos/farmacologia , Quinazolinonas/efeitos adversos , Quinazolinonas/farmacologia , Receptores de Detecção de Cálcio/metabolismo
3.
Eur Respir J ; 52(2)2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29946002

RESUMO

Chronic mucus hypersecretion (CMH) contributes to the morbidity and mortality of asthma, and remains uncontrolled by current therapies in the subset of patients with severe, steroid-resistant disease. Altered cross-talk between airway epithelium and airway smooth muscle cells (ASMCs), driven by pro-inflammatory cytokines such as interleukin (IL)-1ß, provides a potential mechanism that influences CMH. This study investigated mechanisms underlying CMH by comparing IL-1ß-induced gene expression profiles between asthma and control-derived ASMCs and the subsequent paracrine influence on airway epithelial mucus production in vitroIL-1ß-treated ASMCs from asthmatic patients and healthy donors were profiled using microarray analysis and ELISA. Air-liquid interface (ALI)-cultured CALU-3 and primary airway epithelial cells were treated with identified candidates and mucus production assessed.The IL-1ß-induced CCL20 expression and protein release was increased in ASMCs from moderate compared with mild asthmatic patients and healthy controls. IL-1ß induced lower MIR146A expression in asthma-derived ASMCs compared with controls. Decreased MIR146A expression was validated in vivo in bronchial biopsies from 16 asthmatic patients versus 39 healthy donors. miR-146a-5p overexpression abrogated CCL20 release in ASMCs. CCL20 treatment of ALI-cultured CALU-3 and primary airway epithelial cells induced mucus production, while CCL20 levels in sputum were associated with increased levels of CMH in asthmatic patients.Elevated CCL20 production by ASMCs, possibly resulting from dysregulated expression of the anti-inflammatory miR-146a-5p, may contribute to enhanced mucus production in asthma.


Assuntos
Asma/metabolismo , Quimiocina CCL20/metabolismo , Interleucina-1beta/farmacologia , MicroRNAs/metabolismo , Miócitos de Músculo Liso/metabolismo , Adolescente , Adulto , Idoso , Asma/tratamento farmacológico , Estudos de Casos e Controles , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Muco/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Escarro/metabolismo , Adulto Jovem
4.
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
5.
J Physiol ; 596(1): 47-66, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29071730

RESUMO

KEY POINTS: Transforming growth-factor-ß (TGF-ß) and RhoA/Rho-kinase are independently implicated in the airway hyper-responsiveness associated with asthma, but how these proteins interact is not fully understood. We examined the effects of pre-treatment with TGF-ß on expression and activity of RhoA, Rho-kinase and ARHGEF1, an activator of RhoA, as well as on bradykinin-induced contraction, in airway smooth muscle. TGF-ß enhanced bradykinin-induced RhoA translocation, Rho-kinase-dependent phosphorylation and contraction, but partially suppressed bradykinin-induced RhoA activity (RhoA-GTP content). TGF-ß enhanced the expression of ARHGEF1, while a small interfering RNA against ARHGEF1 and a RhoGEF inhibitor prevented the effects of TGF-ß on RhoA and Rho-kinase activity and contraction, respectively. ARHGEF1 expression was also enhanced in airway smooth muscle from asthmatic patients and ovalbumin-sensitized mice. ARHGEF1 is a key TGF-ß target gene, an important regulator of Rho-kinase activity and therefore a potential therapeutic target for the treatment of asthmatic airway hyper-responsiveness. ABSTRACT: Transforming growth factor-ß (TGF-ß), RhoA/Rho-kinase and Src-family kinases (SrcFK) have independently been implicated in airway hyper-responsiveness, but how they interact to regulate airway smooth muscle contractility is not fully understood. We found that TGF-ß pre-treatment enhanced acute contractile responses to bradykinin (BK) in isolated rat bronchioles, and inhibitors of RhoGEFs (Y16) and Rho-kinase (Y27632), but not the SrcFK inhibitor PP2, prevented this enhancement. In cultured human airway smooth muscle cells (hASMCs), TGF-ß pre-treatment enhanced the protein expression of the Rho guanine nucleotide exchange factor ARHGEF1, MLC20 , MYPT-1 and the actin-severing protein cofilin, but not of RhoA, ROCK2 or c-Src. In hASMCs, acute treatment with BK triggered subcellular translocation of ARHGEF1 and RhoA and enhanced auto-phosphorylation of SrcFK and phosphorylation of MYPT1 and MLC20 , but induced de-phosphorylation of cofilin. TGF-ß pre-treatment amplified the effects of BK on RhoA translocation and MYPT1/MLC20 phosphorylation, but suppressed the effects of BK on RhoA-GTP content, SrcFK auto-phosphorylation and cofilin de-phosphorylation. In hASMCs, an ARHGEF1 small interfering RNA suppressed the effects of BK and TGF-ß on RhoA-GTP content, RhoA translocation and MYPT1 and MLC20 phosphorylation, but minimally influenced the effects of TGF-ß on cofilin expression and phosphorylation. ARHGEF1 expression was also enhanced in ASMCs of asthmatic patients and in lungs of ovalbumin-sensitized mice. Our data indicate that TGF-ß enhances BK-induced contraction, RhoA translocation and Rho-kinase activity in airway smooth muscle largely via ARHGEF1, but independently of SrcFK and total RhoA-GTP content. A role for smooth muscle ARHGEF1 in asthmatic airway hyper-responsiveness is worthy of further investigation.


Assuntos
Asma/fisiopatologia , Contração Muscular , Músculo Liso/fisiologia , Sistema Respiratório/fisiopatologia , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Proteína rhoA de Ligação ao GTP/metabolismo , Adulto , Animais , Asma/tratamento farmacológico , Asma/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Músculo Liso/citologia , Músculo Liso/efeitos dos fármacos , Fosforilação , Ratos , Ratos Wistar , Sistema Respiratório/citologia , Sistema Respiratório/efeitos dos fármacos , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Adulto Jovem , Proteína rhoA de Ligação ao GTP/genética
6.
Adv Exp Med Biol ; 967: 335-342, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29047097

RESUMO

Oxidant stress is strongly associated with cardiovascular disease, including pulmonary hypertension, but antioxidant therapies have so far proven ineffective. This is partly due to a lack of understanding of the key role played by reactive oxygen species (ROS) in physiological cell signalling, and partly to the complex interrelationships between generators of ROS (e.g. mitochondria and NADPH oxidases, NOX), cellular antioxidant systems and indeed Ca2+ signalling. At physiological levels ROS reversibly affect the function of numerous enzymes and transcription factors, most often via oxidation of specific protein thiols. Importantly, they also affect pathways that promote ROS generation by NOX or mitochondria (ROS-induced ROS release), which has an inherent propensity for positive feedback and uncontrolled oxidant production. The reason this does not occur under normal conditions reflects in part a high level of compartmentalisation of ROS signalling within the cell, akin to that for Ca2+. This article considers the physiological processes which regulate NOX and mitochondrial ROS production and degradation and their interactions with each other and Ca2+ signalling pathways, and discusses how loss of spatiotemporal constraints and activation of positive feedback pathways may impact on their dysregulation in pulmonary hypertension.


Assuntos
Hipertensão Pulmonar/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Animais , Antioxidantes/metabolismo , Cálcio/metabolismo , Humanos , Hipertensão Pulmonar/fisiopatologia , Mitocôndrias/metabolismo , NADPH Oxidases/metabolismo , Oxirredução
7.
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
8.
Exp Physiol ; 102(9): 1078-1082, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28004868

RESUMO

NEW FINDINGS: What is the topic of this review? The review concerns the role of reactive oxygen species as physiological second messengers in potentiating G-protein-coupled receptor-mediated vasoconstriction and its potential dysregulation by oxidant stress in pulmonary hypertension. What advances does it highlight? The review highlights the concept that physiological signalling by reactive oxygen species must normally be highly compartmentalized to prevent self-regenerating oxidant stress and promiscuous and uncontrolled signalling, which contribute to the aetiology. Pulmonary hypertension is associated with oxidant stress and increased generation of reactive oxygen species (ROS) by NADPH oxidases (NOX), mitochondria and other sources. There is considerable evidence that these contribute to the aetiology via promotion of pulmonary vascular remodelling, endothelial dysfunction and enhanced vasoreactivity. However, it is now recognized that ROS act as important signalling mediators and second messengers in normal physiological conditions. Many ion channels and protein kinases crucial to pulmonary vascular function are directly or indirectly affected by redox/ROS, including K+ , Ca2+ and non-selective cation channels and Rho kinase. However, the inherent difficulties in quantifying ROS, particularly in subcellular compartments, make it uncertain whether these reported effects are of relevance in physiological rather than pathological conditions. In an attempt to address such issues, we have focused on the role of physiologically generated ROS in the regulation of G-protein-coupled receptor (GPCR)-activated vasoconstrictor pathways. We have recently reported a novel mechanism whereby low concentrations of GPCR-linked vasoconstrictors greatly potentiate Ca2+ entry via a NOX1- and ROS-mediated pathway parallel to the classical vasoconstrictor pathways of Ca2+ mobilization and activation of Rho kinase. Our findings imply that ROS signalling is highly compartmentalized in physiological conditions, but that this may be compromised by pathological increases in oxidant production, for example in pulmonary hypertension, leading to promiscuous actions that contribute to the aetiology. This model is consistent with the proposal that targeted antioxidants could prove to be an effective therapy for pulmonary hypertension.


Assuntos
Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/fisiopatologia , Canais Iônicos/metabolismo , Transdução de Sinais/fisiologia , Humanos , Oxirredução , Espécies Reativas de Oxigênio/metabolismo
9.
Br J Pharmacol ; 172(22): 5265-80, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26294392

RESUMO

BACKGROUND AND PURPOSE: The importance of tyrosine kinases in airway smooth muscle (ASM) contraction is not fully understood. The aim of this study was to investigate the role of Src-family kinases (SrcFK) and focal adhesion kinase (FAK) in GPCR-mediated ASM contraction and associated signalling events. EXPERIMENTAL APPROACH: Contraction was recorded in intact or α-toxin permeabilized rat bronchioles. Phosphorylation of SrcFK, FAK, myosin light-chain-20 (MLC20 ) and myosin phosphatase targeting subunit-1 (MYPT-1) was evaluated in cultured human ASM cells (hASMC). [Ca(2+) ]i was evaluated in Fura-2 loaded hASMC. Responses to carbachol (CCh) and bradykinin (BK) and the contribution of SrcFK and FAK to these responses were determined. KEY RESULTS: Contractile responses in intact bronchioles were inhibited by antagonists of SrcFK, FAK and Rho-kinase, while after α-toxin permeabilization, they were sensitive to inhibition of SrcFK and Rho-kinase, but not FAK. CCh and BK increased phosphorylation of MYPT-1 and MLC20 and auto-phosphorylation of SrcFK and FAK. MYPT-1 phosphorylation was sensitive to inhibition of Rho-kinase and SrcFK, but not FAK. Contraction induced by SR Ca(2+) depletion and equivalent [Ca(2+) ]i responses in hASMC were sensitive to inhibition of both SrcFK and FAK, while depolarization-induced contraction was sensitive to FAK inhibition only. SrcFK auto-phosphorylation was partially FAK-dependent, while FAK auto-phosphorylation was SrcFK-independent. CONCLUSIONS AND IMPLICATIONS: SrcFK mediates Ca(2+) -sensitization in ASM, while SrcFK and FAK together and individually influence multiple Ca(2+) influx pathways. Tyrosine phosphorylation is therefore a key upstream signalling event in ASM contraction and may be a viable target for modulating ASM tone in respiratory disease.


Assuntos
Bronquíolos/fisiologia , Cálcio/fisiologia , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Músculo Liso/fisiologia , Quinases Associadas a rho/fisiologia , Quinases da Família src/fisiologia , Adulto , Animais , Bradicinina/farmacologia , Bronquíolos/citologia , Broncoconstritores/farmacologia , Carbacol/farmacologia , Células Cultivadas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Liso/citologia , Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , Ratos Wistar , Traqueia/efeitos dos fármacos , Traqueia/fisiologia , Adulto Jovem
10.
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
11.
J Physiol ; 593(2): 385-401, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25630260

RESUMO

An increase in the H2 S (hydrogen sulphide, hereafter sulphide) concentration in pulmonary artery smooth muscle cells (PASMCs) has been proposed to mediate hypoxic pulmonary vasoconstriction (HPV). We evaluated this hypothesis in isolated rat intrapulmonary arteries (IPAs) by examining the effects of the sulphide precursor cysteine and sulphide-synthesis blockers on HPV and also on normoxic pulmonary vasoconstriction (NPV) stimulated by prostaglandin F2α (PGF2α ) and by the drug LY83583, which causes contraction in IPAs by increasing cellular reactive oxygen species levels. Experiments with several blockers of cystathionine γ-lyase (CSE), the enzyme responsible for sulphide synthesis in the vasculature, demonstrated that propargylglycine (PAG, 1 mm) had little or no effect on the NPV caused by PGF2α or LY83583. Conversely, other CSE antagonists tested, aminooxyacetic acid (AOAA, 100 µm), ß-cyanoalanine (BCA, 500 µm) and hydroxylamine (HA, 100 µm), altered the NPV to PGF2α (BCA increased, HA inhibited) and/or LY83583 (BCA increased, AOAA and HA inhibited). Preincubating IPAs in physiological saline solution (PSS) containing 1 mm cysteine increased the amplitude of the NPV to PGF2(α) by ∼50%, and had a similar effect on HPV elicited by hypoxic challenge with 0% O2 . The enhancement of both responses by cysteine was abolished by pretreatment with 1 mm PAG. Measurements carried out with an amperometric electrode demonstrated that incubation with 1 mm cysteine under anoxic conditions (to minimize sulphide oxidation) greatly potentiated the release of sulphide from pieces of rat liver and that this release was strongly antagonized by PAG, indicating that at this concentration PAG could enter cells intact and antagonize CSE. PAG at 1 mm had no effect on HPV recorded in control PSS, or in PSS supplemented with physiological concentrations of cysteine (10 µm), cystine (50 µm) and glutamate (100 µm) in order to prevent the possible depletion of intracellular cysteine during experiments. Application of a combination of 1 mm cysteine and 1 mm α-ketoglutarate to promote sulphide synthesis via the cysteine aminotransferase/mercaptopyruvate sulphurtransferase (CAT/MST) pathway caused an increase in HPV similar to that observed for cysteine. This was partially blocked by the CAT antagonist aspartate (1 mm) and also by PAG. However, HPV was not increased by 1 mm α-ketoglutarate alone, and HPV in the absence of α-ketoglutarate and cysteine was not attenuated by aspartate. Pretreatment of IPAs with dithiothreitol (DTT, 1 mm), proposed to promote the conversion of mitochondrial thiosulphate to sulphide, did not increase the release of sulphide from pieces of rat liver in either the presence or the absence of 1 mm cysteine, and virtually abolished HPV. The results provide evidence that the sulphide precursor cysteine can promote both NPV and HPV in rat IPA by generating sulphide via a PAG-sensitive pathway, presumably CSE. However, HPV evoked under control conditions was unaffected by the blockade of CSE. Moreover, HPV was not affected by the CAT antagonist aspartate and was blocked rather than enhanced by DTT. The data therefore indicate that sulphide generated by CSE or CAT/MST or from thiosulphate is unlikely to contribute to O2 sensing during HPV in these arteries.


Assuntos
Cistationina gama-Liase/antagonistas & inibidores , Sulfeto de Hidrogênio/metabolismo , Hipóxia/metabolismo , Artéria Pulmonar/metabolismo , Sulfurtransferases/antagonistas & inibidores , Vasoconstrição , Animais , Cisteína/farmacologia , Dinoprosta/farmacologia , Inibidores Enzimáticos/farmacologia , Hipóxia/fisiopatologia , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiologia , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/fisiopatologia , Ratos , Ratos Wistar
12.
Cardiovasc Res ; 104(1): 116-26, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25062958

RESUMO

AIMS: ß-catenin has been shown to be regulated by inducible nitric oxide synthase (NOS) in endothelial cells. We investigated here whether ß-catenin interacts with and regulates endothelial NOS (eNOS) and whether eNOS activation promotes ß-catenin signalling. METHODS AND RESULTS: We identified ß-catenin as a novel eNOS binding protein in human umbilical vein endothelial cells (HUVECs) by mass spectroscopy and western blot analyses of ß-catenin and eNOS immunoprecipitates. This was confirmed by in situ proximity ligation assay. eNOS activity, assessed by cGMP production and eNOS phosphorylation (Ser1177), was enhanced in ß-catenin(-/-) mouse pulmonary endothelial cells (MPECs) relative to wild-type MPECs. eNOS activation (using adenosine, salbutamol, thrombin, or histamine), or application of an NO donor (spermine NONOate) or cGMP-analogue (8-bromo-cGMP) caused nuclear translocation of ß-catenin in HUVEC as shown by western blotting of nuclear extracts. Exposure to spermine NONOate, 8-bromo-cGMP, or sildenafil (a phosphodiesterase type 5 inhibitor) also increased the expression of ß-catenin-dependent transcripts, IL-8, and cyclin D1. Stimulation of wild-type MPECs with basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), spermine NONOate, 8-bromo-cGMP, or sildenafil increased tube length relative to controls in an angiogenesis assay. These responses were abrogated in ß-catenin(-/-) MPECs, with the exception of that to bFGF which is NO-independent. In C57BL/6 mice, subcutaneous VEGF-supplemented Matrigel plugs containing ß-catenin(-/-) MPECs exhibited reduced angiogenesis compared with plugs containing wild-type MPECs. Angiogenesis was not altered in bFGF-supplemented Matrigel. CONCLUSION: These data reveal bidirectional cross-talk and regulation between the NO-cGMP and ß-catenin signalling pathways.


Assuntos
Células Endoteliais da Veia Umbilical Humana/enzimologia , Neovascularização Fisiológica , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , beta Catenina/metabolismo , Animais , Células Cultivadas , GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Quinase 3 da Glicogênio Sintase , Glicogênio Sintase Quinase 3 beta , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Doadores de Óxido Nítrico/farmacologia , Inibidores da Fosfodiesterase 5/farmacologia , Fosforilação , Ligação Proteica , Serina , Transdução de Sinais , beta Catenina/deficiência , beta Catenina/genética
13.
J Physiol ; 591(18): 4473-98, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23774281

RESUMO

Hypoxic pulmonary vasoconstriction (HPV) maintains blood oxygenation during acute hypoxia but contributes to pulmonary hypertension during chronic hypoxia. The mechanisms of HPV remain controversial, in part because HPV is usually studied in the presence of agonist-induced preconstriction ('pretone'). This potentiates HPV but may obscure and distort its underlying mechanisms. We therefore carried out an extensive assessment of proposed mechanisms contributing to HPV in isolated intrapulmonary arteries (IPAs) in the absence of pretone by using a conventional small vessel myograph. Hypoxia elicited a biphasic constriction consisting of a small transient (phase 1) superimposed upon a sustained (phase 2) component. Neither phase was affected by the L-type Ca2+ channel antagonists diltiazem (10 and 30 µm) or nifedipine (3 µm). Application of the store-operated Ca2+ entry (SOCE) blockers BTP2 (10 µm) or SKF96365 (50 µm) attenuated phase 2 but not phase 1, whereas a lengthy (30 min) incubation in Ca2+-free physiological saline solution similarly reduced phase 2 but abolished phase 1. No further effect of inhibition of HPV was observed if the sarco/endoplasmic reticulum Ca2+-ATPase inhibitor cyclopiazonic acid (30 µm) was also applied during the 30 min incubation in Ca2+-free physiological saline solution. Pretreatment with 10 µm ryanodine and 15 mm caffeine abolished both phases, whereas treatment with 100 µm ryanodine attenuated both phases. The two-pore channel blocker NED-19 (1 µm) and the nicotinic acid adenine dinucleotide phosphate (NAADP) antagonist BZ194 (200 µm) had no effect on either phase of HPV. The lysosomal Ca2+-depleting agent concanamycin (1 µm) enhanced HPV if applied during hypoxia, but had no effect on HPV during a subsequent hypoxic challenge. The cyclic ADP ribose antagonist 8-bromo-cyclic ADP ribose (30 µm) had no effect on either phase of HPV. Neither the Ca2+-sensing receptor (CaSR) blocker NPS2390 (0.1 and 10 µm) nor FK506 (10 µm), a drug which displaces FKBP12.6 from ryanodine receptor 2 (RyR2), had any effect on HPV. HPV was virtually abolished by the rho kinase blocker Y-27632 (1 µm) and attenuated by the protein kinase C inhibitor Gö6983 (3 µm). Hypoxia for 45 min caused a significant increase in the ratio of oxidised to reduced glutathione (GSSG/GSH). HPV was unaffected by the NADPH oxidase inhibitor VAS2870 (10 µm), whereas phase 2 was inhibited but phase 1 was unaffected by the antioxidants ebselen (100 µm) and TEMPOL (3 mm). We conclude that both phases of HPV in this model are mainly dependent on [Ca2+]i release from the sarcoplasmic reticulum. Neither phase of HPV requires voltage-gated Ca2+ entry, but SOCE contributes to phase 2. We can detect no requirement for cyclic ADP ribose, NAADP-dependent lysosomal Ca2+ release, activation of the CaSR, or displacement of FKBP12.6 from RyR2 for either phase of HPV. Sustained HPV is associated with an oxidising shift in the GSSG/GSH redox potential and is inhibited by the antioxidants ebselen and TEMPOL, consistent with the concept that it requires an oxidising shift in the cell redox state or the generation of reactive oxygen species.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio , Cálcio/metabolismo , Hipóxia/metabolismo , Artéria Pulmonar/metabolismo , Vasoconstrição , Animais , Antioxidantes/farmacologia , ATPases Transportadoras de Cálcio/antagonistas & inibidores , Glutationa/metabolismo , Masculino , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/fisiologia , Ratos , Ratos Wistar , Receptores de Detecção de Cálcio/antagonistas & inibidores , Rianodina/farmacologia , Retículo Sarcoplasmático/metabolismo
14.
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
15.
J Neurosci ; 32(48): 17502-13, 2012 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-23197740

RESUMO

Chronic neuropathic pain affects millions of individuals worldwide, is typically long-lasting, and remains poorly treated with existing therapies. Neuropathic pain arising from peripheral nerve lesions is known to be dependent on the emergence of spontaneous and evoked hyperexcitability in damaged nerves. Here, we report that the potassium channel subunit Kv9.1 is expressed in myelinated sensory neurons, but is absent from small unmyelinated neurons. Kv9.1 expression was strongly and rapidly downregulated following axotomy, with a time course that matches the development of spontaneous activity and pain hypersensitivity in animal models. Interestingly, siRNA-mediated knock-down of Kv9.1 in naive rats led to neuropathic pain behaviors. Diminished Kv9.1 function also augmented myelinated sensory neuron excitability, manifested as spontaneous firing, hyper-responsiveness to stimulation, and persistent after-discharge. Intracellular recordings from ex vivo dorsal root ganglion preparations revealed that Kv9.1 knock-down was linked to lowered firing thresholds and increased firing rates under physiologically relevant conditions of extracellular potassium accumulation during prolonged activity. Similar neurophysiological changes were detected in animals subjected to traumatic nerve injury and provide an explanation for neuropathic pain symptoms, including poorly understood conditions such as hyperpathia and paresthesias. In summary, our results demonstrate that Kv9.1 dysfunction leads to spontaneous and evoked neuronal hyperexcitability in myelinated fibers, coupled with development of neuropathic pain behaviors.


Assuntos
Regulação para Baixo , Neuralgia/metabolismo , Traumatismos dos Nervos Periféricos/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Células Receptoras Sensoriais/metabolismo , Animais , Axotomia , Comportamento Animal/fisiologia , Hiperalgesia/etiologia , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Masculino , Fibras Nervosas Mielinizadas/metabolismo , Neuralgia/etiologia , Neuralgia/fisiopatologia , Medição da Dor , Traumatismos dos Nervos Periféricos/complicações , Traumatismos dos Nervos Periféricos/fisiopatologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , RNA Interferente Pequeno , Ratos , Ratos Wistar
16.
Physiol Rev ; 92(1): 367-520, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22298659

RESUMO

It has been known for more than 60 years, and suspected for over 100, that alveolar hypoxia causes pulmonary vasoconstriction by means of mechanisms local to the lung. For the last 20 years, it has been clear that the essential sensor, transduction, and effector mechanisms responsible for hypoxic pulmonary vasoconstriction (HPV) reside in the pulmonary arterial smooth muscle cell. The main focus of this review is the cellular and molecular work performed to clarify these intrinsic mechanisms and to determine how they are facilitated and inhibited by the extrinsic influences of other cells. Because the interaction of intrinsic and extrinsic mechanisms is likely to shape expression of HPV in vivo, we relate results obtained in cells to HPV in more intact preparations, such as intact and isolated lungs and isolated pulmonary vessels. Finally, we evaluate evidence regarding the contribution of HPV to the physiological and pathophysiological processes involved in the transition from fetal to neonatal life, pulmonary gas exchange, high-altitude pulmonary edema, and pulmonary hypertension. Although understanding of HPV has advanced significantly, major areas of ignorance and uncertainty await resolution.


Assuntos
Hipóxia/fisiopatologia , Alvéolos Pulmonares/irrigação sanguínea , Vasoconstrição/fisiologia , Doença da Altitude/fisiopatologia , Comunicação Celular , Humanos , Hipertensão Pulmonar/fisiopatologia , Recém-Nascido , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Alvéolos Pulmonares/embriologia , Alvéolos Pulmonares/crescimento & desenvolvimento , Troca Gasosa Pulmonar/fisiologia
17.
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
18.
Antioxid Redox Signal ; 15(6): 1531-47, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20849377

RESUMO

Reactive oxygen species (ROS) are continuously generated in vascular tissues by various oxidoreductase enzymes. They contribute to normal cell signaling, and modulate vascular smooth muscle tone and endothelial permeability in response to physiological agonists and to various cellular stresses and environmental factors, such as hypoxia. While concentrations of ROS are normally tightly controlled by cellular redox buffer systems, if produced in excess they may contribute to vascular disease. Protein kinases are essential components of most cell signaling pathways, including those involving ROS. The functioning of several members of this highly diverse group of enzymes, which include receptor and nonreceptor tyrosine kinases, protein kinase C, mitogen-activated kinases, and Rho-kinase, are modified by ROS, either through direct oxidative modification or indirectly through modification of associated proteins such as tyrosine phosphatases and monomeric G proteins. In this review, we discuss the molecular mechanisms of redox modification of these proteins, the downstream pathways affected, the often complex interaction between major kinase pathways, and feedback to ROS production itself. We also discuss complicating factors such as differential actions of superoxide anion and hydrogen peroxide, questions concerning concentration dependence, and the significance of signaling microdomains.


Assuntos
Proteínas Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Quinases Associadas a rho/metabolismo , Vasos Sanguíneos/enzimologia , Vasos Sanguíneos/fisiologia , Endotélio Vascular/enzimologia , Endotélio Vascular/fisiologia , Ativação Enzimática , Humanos , Peróxido de Hidrogênio/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiologia , Oxirredução , Estresse Oxidativo , Proteína Quinase C/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Superóxido Dismutase , Superóxidos/metabolismo , Quinases da Família src/metabolismo
20.
Thorax ; 65(6): 547-52, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20522856

RESUMO

Asthma is characterised by airway hyper-responsiveness and remodelling, and there is mounting evidence that alterations in the phenotype of airway smooth muscle (ASM) play a central role in these processes. Although the concept that dysregulation of ASM Ca(2+) homeostasis may underlie at least part of these alterations has been around for many years, it is only relatively recently that the availability of ASM biopsies from subjects with mild and moderate asthma has allowed it to be properly investigated. In this article, critical components of the pathobiology of asthmatic ASM, including contractile function, proliferation, cell migration and secretion of proinflammatory cytokines and chemokines, are reviewed and related to associated changes in ASM Ca(2+) homeostasis. Based on this evidence, it is proposed that a unifying mechanism for the abnormal asthmatic phenotype is dysregulation of Ca(2+) homeostasis caused at least in part by a downregulation in expression and function of sarcoendoplasmic Ca(2+) ATPases (SERCAs).


Assuntos
Remodelação das Vias Aéreas/fisiologia , Asma/fisiopatologia , Cálcio/metabolismo , Músculo Liso/fisiopatologia , Citocinas/metabolismo , Homeostase/fisiologia , Humanos , Contração Muscular/fisiologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...