Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cell Commun Signal ; 13: 32, 2015 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-26177720

RESUMO

BACKGROUND: Orai/CRACM1 ion channels provide the major Ca(2+) influx pathway for FcεRI-dependent human lung mast cell (HLMC) mediator release. The Ca(2+)-activated K(+) channel KCa3.1 modulates Ca(2+) influx and the secretory response through hyperpolarisation of the plasma membrane. We hypothesised that there is a close functional and spatiotemporal interaction between these Ca(2+)- and K(+)-selective channels. RESULTS: Activation of FcεRI-dependent HLMC KCa3.1 currents was dependent on the presence of extracellular Ca(2+), and attenuated in the presence of the selective Orai blocker GSK-7975A. Currents elicited by the KCa3.1 opener 1-EBIO were also attenuated by GSK-7975A. The Orai1 E106Q dominant-negative mutant ablated 1-EBIO and FcεRI-dependent KCa3.1 currents in HLMCs. Orai1 but not Orai2 was shown to co-immunoprecipitate with KCa3.1 when overexpressed in HEK293 cells, and Orai1 and KCa3.1 were seen to co-localise in the HEK293 plasma membrane using confocal microscopy. CONCLUSION: KCa3.1 activation in HLMCs is highly dependent on Ca(2+) influx through Orai1 channels, mediated via a close spatiotemporal interaction between the two channels.


Assuntos
Canais de Cálcio/metabolismo , Membrana Celular/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Mastócitos/metabolismo , Cálcio/metabolismo , Canais de Cálcio/análise , Canais de Cálcio/genética , Células Cultivadas , Células HEK293 , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/análise , Pulmão/citologia , Mastócitos/citologia , Proteína ORAI1 , Mutação Puntual , Mapas de Interação de Proteínas
2.
J Immunol ; 191(5): 2624-2636, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23904164

RESUMO

Identifying the factors responsible for relative glucocorticosteroid (GC) resistance present in patients with severe asthma and finding tools to reverse it are of paramount importance. In asthma we see in vivo evidence of GC-resistant pathways in airway smooth muscle (ASM) bundles that can be modeled in vitro by exposing cultured ASM cells to TNF-α/IFN-γ. This action drives GC insensitivity via protein phosphatase 5-dependent impairment of GC receptor phosphorylation. In this study, we investigated whether KCa3.1 ion channels modulate the activity of GC-resistant pathways using our ASM model of GC insensitivity. Immunohistochemical staining of endobronchial biopsies revealed that KCa3.1 channels are localized to the plasma membrane and nucleus of ASM in both healthy controls and asthmatic patients, irrespective of disease severity. Western blot assays and immunofluorescence staining confirmed the nuclear localization of KCa3.1 channels in ASM cells. The functional importance of KCa3.1 channels in the regulation of GC-resistant chemokines induced by TNF-α/IFN-γ was assessed using complementary inhibitory strategies, including KCa3.1 blockers (TRAM-34 and ICA-17043) or KCa3.1-specific small hairpin RNA delivered by adenoviruses. KCa3.1 channel blockade led to a significant reduction of fluticasone-resistant CX3CL1, CCL5, and CCL11 gene and protein expression. KCa3.1 channel blockade also restored fluticasone-induced GC receptor-α phosphorylation at Ser(211) and transactivation properties via the suppression of cytokine-induced protein phosphatase 5 expression. The effect of KCa3.1 blockade was evident in ASM cells from both healthy controls and asthmatic subjects. In summary, KCa3.1 channels contribute to the regulation of GC-resistant inflammatory pathways in ASM cells: blocking KCa3.1 channels may enhance corticosteroid activity in severe asthma.


Assuntos
Asma/metabolismo , Brônquios/metabolismo , Glucocorticoides , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Miócitos de Músculo Liso/metabolismo , Adulto , Asma/tratamento farmacológico , Western Blotting , Células Cultivadas , Resistência a Medicamentos/fisiologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Imunofluorescência , Humanos , Imuno-Histoquímica , Masculino , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase Via Transcriptase Reversa
3.
J Allergy Clin Immunol ; 129(6): 1628-35.e2, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22409987

RESUMO

BACKGROUND: Influx of extracellular Ca(2+) into human lung mast cells (HLMCs) is essential for the FcεRI-dependent release of preformed granule-derived mediators and newly synthesized autacoids and cytokines. However, the identity of the ion channels underlying this Ca(2+) influx is unknown. The recently discovered members of the CRACM/Orai ion channel family that carries the Ca(2+) release-activated Ca(2+) current are candidates. OBJECTIVES: To investigate the expression and function of CRACM channels in HLMCs. METHODS: CRACM mRNA, protein, and functional expression were examined in purified HLMCs and isolated human bronchus. RESULTS: CRACM1, -2, and -3 mRNA transcripts and CRACM1 and -2 proteins were detectable in HLMCs. A CRACM-like current was detected following FcεRI-dependent HLMC activation and also in HLMCs dialyzed with 30 µM inositol triphosphate. The Ca(2+)-selective current obtained under both conditions was blocked by 10 µM La(3+) and Gd(3+), known blockers of CRACM channels, and 2 distinct and specific CRACM-channel blockers-GSK-7975A and Synta-66. Both blockers reduced FcεRI-dependent Ca(2+) influx, and 3 µM GSK-7975A and Synta-66 reduced the release of histamine, leukotriene C(4), and cytokines (IL-5/-8/-13 and TNFα) by up to 50%. Synta-66 also inhibited allergen-dependent bronchial smooth muscle contraction in ex vivo tissue. CONCLUSIONS: The presence of CRACM channels, a CRACM-like current, and functional inhibition of HLMC Ca(2+) influx, mediator release, and allergen-induced bronchial smooth muscle contraction by CRACM-channel blockers supports a role for CRACM channels in FcεRI-dependent HLMC secretion. CRACM channels are therefore a potential therapeutic target in the treatment of asthma and related allergic diseases.


Assuntos
Canais de Cálcio/metabolismo , Pulmão/metabolismo , Mastócitos/metabolismo , Alérgenos/imunologia , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/genética , Linhagem Celular , Humanos , Imunoglobulina E/metabolismo , Pulmão/citologia , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Músculo Liso/metabolismo , RNA Mensageiro/metabolismo
4.
J Allergy Clin Immunol ; 128(6): 1303-1309.e2, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21872912

RESUMO

BACKGROUND: Fibrocytes are bone marrow-derived CD34(+) collagen I-positive cells present in peripheral blood that develop α-smooth muscle actin expression and contractile activity in tissue culture. They are implicated in the pathogenesis of tissue remodeling and fibrosis in both patients with asthma and those with idiopathic pulmonary fibrosis. Targeting fibrocyte migration might therefore offer a new approach for the treatment of these diseases. Ion channels play key roles in cell function, but the ion-channel repertoire of human fibrocytes is unknown. OBJECTIVE: We sought to examine whether human fibrocytes express the K(Ca)3.1 K(+) channel and to determine its role in cell differentiation, survival, and migration. METHODS: Fibrocytes were cultured from the peripheral blood of healthy subjects and patients with asthma. Whole-cell patch-clamp electrophysiology was used for the measurement of ion currents, whereas mRNA and protein were examined to confirm channel expression. Fibrocyte migration and proliferation assays were performed in the presence of K(Ca)3.1 ion-channel blockers. RESULTS: Human fibrocytes cultured from the peripheral blood of both healthy control subjects and asthmatic patients expressed robust K(Ca)3.1 ion currents together with K(Ca)3.1 mRNA and protein. Two specific and distinct K(Ca)3.1 blockers (TRAM-34 and ICA-17043) markedly inhibited fibrocyte migration in transwell migration assays. Channel blockers had no effect on fibrocyte growth, apoptosis, or differentiation in cell culture. CONCLUSIONS: The K(+) channel K(Ca)3.1 plays a key role in human fibrocyte migration. Currently available K(Ca)3.1-channel blockers might therefore attenuate tissue fibrosis and remodeling in patients with diseases such as idiopathic pulmonary fibrosis and asthma through the inhibition of fibrocyte recruitment.


Assuntos
Asma/metabolismo , Movimento Celular , Células do Tecido Conjuntivo/citologia , Células do Tecido Conjuntivo/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Remodelação das Vias Aéreas/fisiologia , Asma/patologia , Western Blotting , Diferenciação Celular/fisiologia , Fibrose/metabolismo , Fibrose/patologia , Humanos , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
J Allergy Clin Immunol ; 125(1): 257-63.e1-5, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19864009

RESUMO

BACKGROUND: Mast cells contribute to the pathophysiology of asthma with the sustained release of both preformed and newly generated mediators in response to allergens and other diverse stimuli. Stem cell factor (SCF) is the key human mast cell growth factor, but also primes mast cells for mediator release. SCF expression is markedly increased in asthmatic airways. Short-acting beta(2)-adrenoceptor drugs such as albuterol inhibit human lung mast cell (HLMC) degranulation in vitro in the absence of SCF, but their effect in the presence of SCF is not known. OBJECTIVE: The aim of this study was to elucidate the effects of albuterol on HLMC function in the presence of SCF. METHODS: Mediator release and K(Ca)3.1 ion channel activity were analyzed in purified HLMC. Intracellular signalling and beta(2)-adrenoceptor phosphorylation and internalization were analyzed in the HMC-1 human mast cell line. RESULTS: beta(2)-Adrenoceptor agonist-dependent inhibition of K(Ca)3.1 ion channels and HLMC mediator release was markedly attenuated in the presence of SCF. Remarkably, albuterol actually potentiated IgE-induced histamine release in a dose-dependent manner when both SCF and IgE were present. These effects were related to the SCF-dependent phosphorylation of Tyr350 on the beta(2)-adrenoceptor with immediate uncoupling of the receptor followed by receptor internalization. CONCLUSION: The potentially beneficial effects of beta(2)-adrenoceptor agonists in asthmatic airways may be blunted as a result of the high concentrations of SCF present.


Assuntos
Albuterol , Pulmão , Mastócitos/efeitos dos fármacos , Mastócitos/metabolismo , Receptores Adrenérgicos beta 2 , Fator de Células-Tronco/farmacologia , Albuterol/metabolismo , Albuterol/farmacologia , Asma/tratamento farmacológico , Asma/metabolismo , Asma/fisiopatologia , Linhagem Celular , Liberação de Histamina , Humanos , Pulmão/citologia , Pulmão/efeitos dos fármacos , Mastócitos/imunologia , Canais de Potássio Cálcio-Ativados/metabolismo , Receptores Adrenérgicos beta 2/efeitos dos fármacos , Receptores Adrenérgicos beta 2/metabolismo , Fator de Células-Tronco/metabolismo
6.
Br J Pharmacol ; 178(15): 2948-2962, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33786825

RESUMO

BACKGROUND AND PURPOSE: TGFß1-mediated myofibroblast activation contributes to pathological fibrosis in many diseases including idiopathic pulmonary fibrosis (IPF), where myofibroblast resistance to oxidant-mediated apoptosis is also evident. We therefore investigated the involvement of redox-sensitive TRPA1 ion channels on human lung myofibroblasts (HLMFs) cell death and TGFß1-mediated pro-fibrotic responses. EXPERIMENTAL APPROACH: The effects of TGFß1 stimulation on TRPA1 expression and cell viability was studied in HLMFs derived from IPF patients and non-fibrotic patients. We also examined a model of TGFß1-dependent fibrogenesis in human lung. We used qRT-PCR, immunofluorescent assays, overexpression with lentiviral vectors and electrophysiological methods. KEY RESULTS: TRPA1 mRNA, protein and ion currents were expressed in HLMFs derived from both non-fibrotic patient controls and IPF patients, and expression was reduced by TGFß1. TRPA1 mRNA was also down-regulated by TGFß1 in a model of lung fibrogenesis in human lung. TRPA1 over-expression or activation induced HLMF apoptosis, and activation of TRPA1 channel activation by H2 O2 induced necrosis. TRPA1 inhibition following TGFß1 down-regulation or pharmacological inhibition, protected HLMFs from both apoptosis and necrosis. Lentiviral vector mediated TRPA1 expression was also found to induce sensitivity to H2 O2 induced cell death in a TRPA1-negative HEK293T cell line. CONCLUSION AND IMPLICATIONS: TGFß1 induces resistance of HLMFs to TRPA1 agonist- and H2 O2 -mediated cell death via down-regulation of TRPA1 channels. Our data suggest that therapeutic strategies which prevent TGFß1-dependent down-regulation of TRPA1 may reduce myofibroblast survival in IPF and therefore improve clinical outcomes.


Assuntos
Miofibroblastos , Canal de Cátion TRPA1 , Fator de Crescimento Transformador beta1 , Apoptose , Regulação para Baixo , Fibroblastos/metabolismo , Células HEK293 , Humanos , Pulmão/metabolismo , Miofibroblastos/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
7.
Am J Respir Cell Mol Biol ; 37(5): 525-31, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17585114

RESUMO

Airway smooth muscle cell hyperplasia contributes to airway remodeling and hyperreactivity characteristic of asthma. Changes to potassium channel activity in proliferating human airway smooth muscle (HASM) cells have been described, but no regulatory role in proliferation has been attributed to them. We sought to investigate the expression of the intermediate conductance calcium-activated potassium channel K(Ca)3.1 in HASM cells and investigate its role in proliferation. Smooth muscle cells derived from human airways were grown in vitro and K(Ca)3.1 channel expression was measured using Western blot, RT-PCR, and patch clamp electrophysiology. Pharmacologic inhibitors of the channel were used in assays of cellular proliferation, and flow cytometry was used to identify cell cycle regulation. HASM cells expressed K(Ca)3.1 channel mRNA, protein, and activity with up-regulation evident after transforming growth factor-beta stimulation. Pharmacologic inhibition of K(Ca)3.1 led to growth arrest in cells stimulated to proliferate with mitogens. These inhibitors did not cause cellular toxicity or induce apoptosis. We have demonstrated, for the first time, the expression of K(Ca)3.1 channels in HASM cells. In addition, we have shown that K(Ca)3.1 channels are important in HASM cell proliferation, making these channels a potential therapeutic target in airway remodeling.


Assuntos
Cálcio/fisiologia , Proliferação de Células , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/fisiologia , Músculo Liso/citologia , Potássio/metabolismo , Sistema Respiratório/citologia , Células Cultivadas , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/biossíntese , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Músculo Liso/metabolismo , RNA Mensageiro/metabolismo , Sistema Respiratório/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Regulação para Cima/fisiologia
8.
FASEB J ; 19(8): 1006-8, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15817638

RESUMO

Human mast cells express the intermediate conductance Ca2+-activated K+ channel iKCa1, which opens following IgE-dependent activation. This results in cell membrane hyperpolarization and potentiation of both Ca2+ influx and degranulation. Mast cell activation is attenuated following exposure to beta2-adrenoceptor agonists such as salbutamol, an effect postulated to operate via intracellular cyclic AMP. In this study, we show that salbutamol closes iKCa1 in mast cells derived from human lung and peripheral blood. Salbutamol (1-10 microM) inhibited iKCa1 currents following activation with both anti-IgE and the iKCa1 opener 1-EBIO, and was reversed by removing salbutamol or by the addition of the selective beta2-adrenoceptor antagonist and inverse agonist ICI 118551. Interestingly, ICI 118551 consistently opened iKCa1 in quiescent cells, suggesting that constitutive beta2-receptor signaling suppresses channel activity. Manipulation of intracellular cAMP, Galphai, and Galphas demonstrates that the beta2-adrenergic effects are consistent with a membrane-delimited mechanism involving Galphas. This is the first demonstration that gating of the iKCa1 channel is regulated by a G protein-coupled receptor and provides a clearly defined mechanism for the mast cell "stabilizing" effect of beta2-agonists. Furthermore, the degree of constitutive beta2-receptor "tone" may control the threshold for human mast cell activation through the regulation of iKCa1.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Intermediária/fisiologia , Mastócitos/fisiologia , Receptores Adrenérgicos beta 2/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Albuterol/farmacologia , Anticorpos/farmacologia , Benzimidazóis/farmacologia , Células Cultivadas , Condutividade Elétrica , Subunidades alfa Gs de Proteínas de Ligação ao GTP , Humanos , Imunoglobulina E/imunologia , Imunoglobulina E/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/efeitos dos fármacos , Ativação do Canal Iônico/efeitos dos fármacos , Pulmão/citologia , Mastócitos/química , Mastócitos/efeitos dos fármacos , Propanolaminas/farmacologia
9.
PLoS One ; 10(12): e0145259, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26689552

RESUMO

The KCa3.1 K+ channel has been proposed as a novel target for pulmonary diseases such as asthma and pulmonary fibrosis. It is expressed in epithelia but its expression and function in primary human bronchial epithelial cells (HBECs) has not been described. Due to its proposed roles in the regulation of cell proliferation, migration, and epithelial fluid secretion, inhibiting this channel might have either beneficial or adverse effects on HBEC function. The aim of this study was to assess whether primary HBECs express the KCa3.1 channel and its role in HBEC function. Primary HBECs from the airways of healthy and asthmatic subjects, SV-transformed BEAS-2B cells and the neoplastic H292 epithelial cell line were studied. Primary HBECs, BEAS-2B and H292 cells expressed KCa3.1 mRNA and protein, and robust KCa3.1 ion currents. KCa3.1 protein expression was increased in asthmatic compared to healthy airway epithelium in situ, and KCa3.1 currents were larger in asthmatic compared to healthy HBECs cultured in vitro. Selective KCa3.1 blockers (TRAM-34, ICA-17043) had no effect on epithelial cell proliferation, wound closure, ciliary beat frequency, or mucus secretion. However, several features of TGFß1-dependent epithelial-mesenchymal transition (EMT) were inhibited by KCa3.1 blockade. Treatment with KCa3.1 blockers is likely to be safe with respect to airway epithelial biology, and may potentially inhibit airway remodelling through the inhibition of EMT.


Assuntos
Brônquios/metabolismo , Células Epiteliais/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/biossíntese , Mucosa Respiratória/metabolismo , Asma/metabolismo , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Regulação da Expressão Gênica , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Fator de Crescimento Transformador beta1/metabolismo
10.
PLoS One ; 8(9): e74895, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24040356

RESUMO

BACKGROUND: The influx of extracellular Ca(2+) into mast cells is critical for the FcεR1-dependent release of preformed granule-derived mediators and newly synthesised autacoids and cytokines. The Orai(CRACM) ion channel family provide the major pathway through which this Ca(2+) influx occurs. However the individual role of each of the three members of the Orai channel family in Ca(2+) influx and mediator release has not been defined in human mast cells. OBJECTIVE: To assess whether there might be value in targeting individual Orai family members for the inhibition of FcεRI-dependent human lung mast cells (HLMC) mediator release. METHODS: We used an adenoviral delivery system to transduce HLMCs with shRNAs targeted against Orai1 and Orai2 or with cDNAs directing the expression of dominant-negative mutations of the three known Orai channels. RESULTS: shRNA-mediated knockdown of Orai1 resulted in a significant reduction of approximately 50% in Ca(2+) influx and in the release of ß-hexosaminidase (a marker of degranulation) and newly synthesized LTC4 in activated HLMCs. In contrast shRNA knockdown of Orai2 resulted in only marginal reductions of Ca(2+) influx, degranulation and LTC4 release. Transduced dominant-negative mutants of Orai1, -2 and -3 markedly reduced Orai currents and completely inhibited HLMC degranulation suggesting that Orai channels form heteromultimers in HLMCs, and that Orai channels comprise the dominant Ca(2+) influx pathway following FceRI-dependent HLMC activation. Inhibition of Orai currents did not alter HLMC survival. In addition we observed a significant down-regulation of the level of CRACM3 mRNA transcripts together with a small increase in the level of CRACM1 and CRACM2 transcripts following a period of sustained HLMC activation. CONCLUSION AND CLINICAL RELEVANCE: Orai1 plays an important role in Ca(2+) influx and mediator release from HLMCs. Strategies which target Orai1 will effectively inhibit FcεRI-dependent HLMC activation, but spare off-target inhibition of Orai2 in other cells and body systems.


Assuntos
Canais de Cálcio/fisiologia , Cálcio/metabolismo , Regulação da Expressão Gênica , Pulmão/citologia , Mastócitos/citologia , Proteínas de Membrana/fisiologia , Adenoviridae/metabolismo , Canais de Cálcio/genética , Sobrevivência Celular , DNA Complementar/metabolismo , Humanos , Proteínas de Membrana/genética , Mutação , Proteína ORAI1 , Proteína ORAI2 , Técnicas de Patch-Clamp , RNA Interferente Pequeno/metabolismo , Receptores de IgE/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo
11.
Eur J Immunol ; 38(9): 2548-56, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18792407

RESUMO

Human lung mast cells (HLMC) express the Ca(2+)-activated K(+) channel K(Ca)3.1, which plays a crucial role in their migration to a variety of diverse chemotactic stimuli. K(Ca)3.1 activation is attenuated by the beta(2)-adrenoceptor and the adenosine A(2A) receptor through a G(s)-coupled mechanism independent of cyclic AMP. Prostaglandin E(2) promotes degranulation and migration of mouse bone marrow-derived mast cells through the G(i)-coupled EP(3) prostanoid receptor, and induces LTC(4) and cytokine secretion from human cord blood-derived mast cells. However, PGE(2) binding to the G(s)-coupled EP(2) receptor on HLMC inhibits their degranulation. We show that EP(2) receptor engagement closes K(Ca)3.1 in HLMC. The EP(2) receptor-specific agonist butaprost was more potent than PGE(2) in this respect, and the effects of both agonists were reversed by the EP(2) receptor antagonist AH6809. Butaprost markedly inhibited HLMC migration induced by chemokine-rich airway smooth muscle-conditioned media. Interestingly, PGE(2) alone was chemotactic for HLMC at high concentrations (1 microM), but was a more potent chemoattractant for HLMC following EP(2) receptor blockade. Therefore, the G(s)-coupled EP(2) receptor closes K(Ca)3.1 in HLMC and attenuates both chemokine- and PGE(2)-dependent HLMC migration. EP(2) receptor agonists with K(Ca)3.1 modulating function may be useful for the treatment of mast cell-mediated disease.


Assuntos
Mastócitos/fisiologia , Canais de Potássio/imunologia , Receptores de Prostaglandina E/metabolismo , Alprostadil/análogos & derivados , Alprostadil/farmacologia , Benzimidazóis/farmacologia , Agonistas dos Canais de Cálcio/farmacologia , Movimento Celular/efeitos dos fármacos , Quimiotaxia , Dinoprostona/metabolismo , Liberação de Histamina , Humanos , Mastócitos/efeitos dos fármacos , Mastócitos/imunologia , Canais de Potássio/metabolismo , Antagonistas de Prostaglandina/farmacologia , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP2 , Xantonas/farmacologia
12.
Eur J Immunol ; 37(6): 1653-62, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17474152

RESUMO

Human lung mast cells (HLMC) express the Ca2+-activated K+ channel KCa3.1, which opens following IgE-dependent activation. This hyperpolarises the cell membrane and potentiates both Ca2+ influx and degranulation. In addition, blockade of KCa3.1 profoundly inhibits HLMC migration to a variety of diverse chemotactic stimuli. KCa3.1 activation is attenuated by the beta2adrenoceptor through a Galphas-coupled mechanism independent of cyclic AMP. Adenosine is an important mediator that both attenuates and enhances HLMC mediator release through the Galphas-coupled A2A and A2B adenosine receptors, respectively. We show that at concentrations that inhibit HLMC degranulation (10(-5)-10(-3) M), adenosine closes KCa3.1 both dose-dependently and reversibly. KCa3.1 suppression by adenosine was reversed partially by the selective adenosine A2A receptor antagonist ZM241385 but not by the A2B receptor antagonist MRS1754, and the effects of adenosine were mimicked by the selective A2A receptor agonist CGS21680. Adenosine also opened a depolarising current carried by non-selective cations. As predicted from the role of KCa3.1 in HLMC migration, adenosine abolished HLMC chemotaxis to asthmatic airway smooth muscle-conditioned medium. In summary, the Galphas-coupled adenosine A2A receptor closes KCa3.1, providing a clearly defined mechanism by which adenosine inhibits HLMC migration and degranulation. A2A receptor agonists with channel-modulating function may be useful for the treatment of mast cell-mediated disease.


Assuntos
Adenosina/farmacologia , Movimento Celular/efeitos dos fármacos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Mastócitos/efeitos dos fármacos , Receptor A2A de Adenosina/metabolismo , Adenosina/análogos & derivados , Agonistas do Receptor A2 de Adenosina , Antagonistas do Receptor A2 de Adenosina , Benzimidazóis/farmacologia , Agonistas dos Canais de Cálcio/farmacologia , Cátions/metabolismo , Meios de Cultivo Condicionados/farmacologia , Estimulação Elétrica , Eletrofisiologia , Histamina/metabolismo , Humanos , Imunoglobulina E/farmacologia , Pulmão/citologia , Mastócitos/metabolismo , Mastócitos/fisiologia , Fenetilaminas/farmacologia , Receptor A2B de Adenosina/metabolismo , Triazinas/farmacologia , Triazóis/farmacologia
13.
J Immunol ; 179(6): 4045-52, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17785843

RESUMO

Mast cells play a significant role in the pathophysiology of many diverse diseases such as asthma and pulmonary fibrosis. Ca2+ influx is essential for mast cell degranulation and release of proinflammatory mediators, while Mg2+ plays an important role in cellular homeostasis. The channels supporting divalent cation influx in human mast cells have not been identified, but candidate channels include the transient receptor potential melastatin (TRPM) family. In this study, we have investigated TRPM7 expression and function in primary human lung mast cells (HLMCs) and in the human mast cell lines LAD2 and HMC-1, using RT-PCR, patch clamp electrophysiology, and RNA interference. Whole cell voltage-clamp recordings revealed a nonselective cation current that activated spontaneously following loss of intracellular Mg2+. The current had a nonlinear current-voltage relationship with the characteristic steep outward rectification associated with TRPM7 channels. Reducing external divalent concentration from 3 to 0.3 mM dramatically increased the size of the outward current, whereas the current was markedly inhibited by elevated intracellular Mg2+ (6 mM). Ion substitution experiments revealed cation selectivity and Ca2+ permeability. RT-PCR confirmed the presence of mRNA for TRPM7 in HLMC, LAD2, and HMC-1 cells. Adenoviral-mediated knockdown of TRPM7 in HLMC with short hairpin RNA and in HMC-1 with short interfering RNA markedly reduced TRPM7 currents and induced cell death, an effect that was not rescued by raising extracellular Mg2+. In summary, HLMC and human mast cell lines express the nonselective cation channel TRPM7 whose presence is essential for cell survival.


Assuntos
Mastócitos/citologia , Mastócitos/metabolismo , Potenciais da Membrana/imunologia , Canais de Cátion TRPM/fisiologia , Agonistas de Receptores Adrenérgicos beta 2 , Cátions Bivalentes/metabolismo , Morte Celular/genética , Morte Celular/imunologia , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Humanos , Pulmão/citologia , Pulmão/imunologia , Pulmão/metabolismo , Mastócitos/imunologia , Potenciais da Membrana/genética , Técnicas de Patch-Clamp , Proteínas Serina-Treonina Quinases , Interferência de RNA/imunologia , RNA Mensageiro/biossíntese , Receptores Adrenérgicos beta 2/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Cátion TRPM/biossíntese , Canais de Cátion TRPM/deficiência , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo , Transdução Genética
14.
J Allergy Clin Immunol ; 112(5): 965-72, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14610489

RESUMO

BACKGROUND: Human lung mast cells (HLMCs) and the human mast cell line HMC-1 express a strongly outwardly rectifying Cl- current characteristic of that carried by the voltage-dependent Cl- channel ClC-5. A similar but distinct current has been implicated in the control of cell proliferation in astrocytes. OBJECTIVE: In this study, we have examined the effects of the Cl- channel blocker tamoxifen on ion channel activity and cell proliferation in both HMC-1 and HLMCs. METHODS: We used the whole-cell patch-clamp technique to characterize macroscopic ion currents in mast cells before and after addition of tamoxifen. HMC-1 proliferation was assessed by incorporation of tritiated thymidine, HLMC proliferation was determined by counting cells in long-term culture, and cell viability was assessed by annexin V binding and propidium iodide uptake. RESULTS: In HMC-1, tamoxifen reduced the outward Cl- current at +130 mV by 73% +/- 9% at a concentration of 3 micromol/L and simultaneously opened a novel inwardly rectifying nonselective cation current with a mean inward current of 153 +/- 18 pA at -130 mV. Tamoxifen produced a dose-dependent inhibition of HMC-1 proliferation (90.3% +/- 4.0% inhibition at 30 micromol/L) without altering cell viability. Tamoxifen inhibited the outward ClC-5-like current in HLMCs, did not open an inward current, and produced a dose-dependent inhibition of HLMC proliferation in long-term culture. CONCLUSION: Tamoxifen inhibits HMC proliferation, possibly through ion channel modulation. This suggests that tamoxifen might be useful in the treatment of mast-cell-mediated diseases, including mastocytosis, asthma, and pulmonary fibrosis.


Assuntos
Canais Iônicos/metabolismo , Mastócitos/patologia , Mastócitos/fisiologia , Tamoxifeno/farmacologia , Cálcio/metabolismo , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Condutividade Elétrica , Histamina/metabolismo , Humanos , Leucemia/patologia , Pulmão/citologia , Mastócitos/citologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-kit/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa