Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Br J Pharmacol ; 164(1): 83-92, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21418186

RESUMO

BACKGROUND AND PURPOSE: Lymphangioleiomyomatosis (LAM) is characterized by the abnormal growth of smooth muscle-like cells (LAM cells) and cystic destruction of the lung parenchyma. LAM cell-derived matrix metalloproteinases (MMPs) are thought to play a prominent role in the tissue destruction. The aim of this study was to determine whether doxycycline, a known MMP inhibitor, can inhibit LAM cell proliferation or mitochondrial function and/or modulate MMPs and their tissue inhibitors (TIMPs). EXPERIMENTAL APPROACH: Wild-type and tuberous sclerosis complex-2 (TSC2)-null mouse embryonic fibroblasts (MEFs) were cultured in DMEM containing 10% fetal bovine serum (FBS). Human LAM cells were derived from the lungs of LAM patients and airway smooth muscle cells from control subjects. Cells were stimulated with FBS with or without doxycycline for up to 9 days. Proliferation was assessed by manual cell counts and MTT assay, MMP production by zymography and ELISA, and TIMP production using elisa. KEY RESULTS: Doxycycline did not change FBS-induced proliferation in MEFs or human cells. However, doxycycline did reduce metabolic activity of both wild-type and TSC2-null MEFs and LAM cells, but had no effect on control cells. Furthermore, doxycycline reduced MMP-2 from MEFs and decreased active-MMP-2 from LAM cells but had no effect on TIMP-1 and TIMP-2 from human LAM cells. CONCLUSIONS AND IMPLICATIONS: Doxycycline decreased MMP levels and cell metabolic activity, which raises the possibility of therapeutic efficacy in LAM.


Assuntos
Doxiciclina/farmacologia , Linfangioleiomiomatose/tratamento farmacológico , Linfangioleiomiomatose/metabolismo , Inibidores de Metaloproteinases de Matriz , Proteínas Supressoras de Tumor/deficiência , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Humanos , Linfangioleiomiomatose/patologia , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Pessoa de Meia-Idade , Mitocôndrias/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/metabolismo
2.
Br J Pharmacol ; 158(6): 1429-41, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19814732

RESUMO

BACKGROUND AND PURPOSE: Airway remodelling in asthma is manifested, in part, as increased airway smooth muscle (ASM) mass, reflecting myocyte proliferation. We hypothesized that calcitriol, a secosteroidal vitamin D receptor (VDR) modulator, would inhibit growth factor-induced myocyte proliferation. EXPERIMENTAL APPROACH: Human ASM cell cultures were derived from bronchial samples taken during surgery. ASM cells were treated with platelet-derived growth factor (PDGF) (10 ng.mL(-1)) for 24 h in the presence of calcitriol, dexamethasone or a checkpoint kinase 1 (Chk1) inhibitor (SB218078). The effects of calcitriol on PDGF-mediated cell proliferation were assessed by thymidine incorporation assay, propidium iodide-based cell cycle analysis, caspase-3 assay and immunoblotting for specific cell cycle modulators. KEY RESULTS: Calcitriol, but not dexamethasone, inhibited PDGF-induced ASM DNA synthesis concentration dependently (IC(50)= 520 +/- 52 nM). These effects were associated with VDR-mediated expression of cytochrome CYP24A1 with no effects on ASM apoptosis. Calcitriol substantially inhibited (P < 0.01) PDGF-stimulated cell growth in ASM derived from both normal (59 +/- 8%) and asthmatic subjects (57 +/- 9%). Calcitriol inhibited PDGF-induced phosphorylation of retinoblastoma protein (Rb) and Chk1, with no effects on PDGF-mediated activation of extracellular signal-regulated kinases 1/2, PI3-kinase and S6 kinase, or expression of p21(Waf/Cip-1), p27(Kip1), cyclin D and E2F-1. Consistent with these observations, SB218078 also inhibited (IC(50)= 450 +/- 100 pM) PDGF-induced cell cycle progression. CONCLUSIONS AND IMPLICATIONS: Calcitriol decreased PDGF-induced ASM cell growth by inhibiting Rb and Chk1 phosphorylation.


Assuntos
Remodelação das Vias Aéreas/efeitos dos fármacos , Calcitriol/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , Vitaminas/farmacologia , Adulto , Asma/tratamento farmacológico , Asma/fisiopatologia , Brônquios/citologia , Brônquios/metabolismo , Calcitriol/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Quinase 1 do Ponto de Checagem , Dexametasona/farmacologia , Feminino , Glucocorticoides/farmacologia , Humanos , Concentração Inibidora 50 , Masculino , Pessoa de Meia-Idade , Miócitos de Músculo Liso/metabolismo , Fosforilação/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Quinases/efeitos dos fármacos , Proteínas Quinases/metabolismo , Proteína do Retinoblastoma/efeitos dos fármacos , Proteína do Retinoblastoma/metabolismo , Vitaminas/administração & dosagem
3.
J Biol Chem ; 276(35): 32648-56, 2001 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-11418617

RESUMO

Despite a widely accepted role of arrestins as "uncouplers" of G protein-coupled receptor (GPCR) signaling, few studies have demonstrated the ability of arrestins to affect second messenger generation by endogenously expressed receptors in intact cells. In this study we demonstrate arrestin specificity for endogenous GPCRs in primary cultures of human airway smooth muscle (HASM). Expression of arrestin-green fluorescent protein (ARR2-GFP or ARR3-GFP) chimeras in HASM significantly attenuated isoproterenol (beta(2)-adrenergic receptor (beta(2)AR)-mediated)- and 5'-(N-ethylcarboxamido)adenosine (A2b adenosine receptor-mediated)-stimulated cAMP production, with fluorescent microscopy demonstrating agonist-promoted redistribution of cellular ARR2-GFP into a punctate formation. Conversely, prostaglandin E(2) (PGE(2))-mediated cAMP production was unaffected by arrestin-GFP, and PGE(2) had little effect on arrestin-GFP distribution. The pharmacological profile of various selective EP receptor ligands suggested a predominantly EP2 receptor population in HASM. Further analysis in COS-1 cells revealed that ARR2-GFP expression increased agonist-promoted internalization of wild type beta(2)AR and EP4 receptors, whereas EP2 receptors remained resistant to internalization. However, expression of an arrestin whose binding to GPCRs is largely independent of receptor phosphorylation (ARR2(R169E)-GFP) enabled substantial agonist-promoted EP2 receptor internalization, increased beta(2)AR internalization to a greater extent than did ARR2-GFP, yet promoted EP4 receptor internalization to the same degree as did ARR2-GFP. Signaling via endogenous EP4 receptors in CHO-K1 cells was attenuated by ARR2-GFP expression, whereas ARR2(R169E)-GFP expression in HASM inhibited EP2 receptor-mediated cAMP production. These findings demonstrate differential effects of arrestins in altering endogenous GPCR signaling in a physiologically relevant cell type and reveal a variable dependence on receptor phosphorylation in dictating arrestin-receptor interaction.


Assuntos
Arrestinas/fisiologia , Músculo Liso/fisiologia , Fosfoproteínas/fisiologia , Receptores Adrenérgicos beta 2/fisiologia , Receptores de Prostaglandina E/fisiologia , Receptores Purinérgicos P1/fisiologia , Transdução de Sinais/fisiologia , Traqueia/fisiologia , Adenosina-5'-(N-etilcarboxamida)/farmacologia , Animais , Arrestinas/genética , Células CHO , Células COS , Linhagem Celular , Células Cultivadas , Chlorocebus aethiops , Cricetinae , AMP Cíclico/metabolismo , Dinoprostona/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde , Humanos , Isoproterenol/farmacologia , Cinética , Proteínas Luminescentes/genética , Músculo Liso/citologia , Fosfoproteínas/genética , Fosforilação , Transporte Proteico , Receptores Adrenérgicos beta 2/efeitos dos fármacos , Receptores de Prostaglandina E/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP2 , Receptores Purinérgicos P1/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo , Traqueia/citologia , Transfecção
5.
J Immunol ; 166(1): 155-61, 2001 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11123288

RESUMO

VCAM-1 is a member of the Ig superfamily of receptors the expression of which is up-regulated on human airway smooth muscle (ASM) cells following stimulation with inflammatory mediators. The function of these receptors in adhesion is well known, but there is growing recognition that they also possess "outside-in" signaling functions, such as cytoskeletal reorganization, calcium mobilization, and cytokine release. The present study examined the activation of extracellular signal-regulated kinase and phosphatidylinositol 3-kinase (PI3K) in ASM cells following VCAM-1 engagement. VCAM-1 ligation activated extracellular signal-regulated kinase 2 and resulted in increased expression of cyclin D1, yet there was neither p27(kip1) degradation nor an increase in smooth muscle cell DNA synthesis. VCAM-1 ligation, however, augmented the proliferative response to submitogenic concentrations of epidermal growth factor. VCAM-1 engagement also stimulated a rapid increase in PI3K activity. This was associated with phosphorylation of the adapter protein p120(Cbl) and an increase in Cbl-associated PI3K activity. These studies suggest that VCAM-1 is linked to multiple signaling pathways in human ASM cells and may function to augment growth factor-induced responses.


Assuntos
Músculo Liso/enzimologia , Músculo Liso/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Traqueia/enzimologia , Traqueia/imunologia , Ubiquitina-Proteína Ligases , Molécula 1 de Adesão de Célula Vascular/fisiologia , Divisão Celular/imunologia , Células Cultivadas , Sinergismo Farmacológico , Ativação Enzimática/imunologia , Humanos , Interleucina-4/farmacologia , Ligantes , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Músculo Liso/citologia , Músculo Liso/metabolismo , Fosforilação , Fosfotirosina/metabolismo , Proteínas Proto-Oncogênicas c-cbl , Traqueia/citologia , Traqueia/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Molécula 1 de Adesão de Célula Vascular/biossíntese , Molécula 1 de Adesão de Célula Vascular/imunologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
6.
Am J Respir Cell Mol Biol ; 23(4): 546-54, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11017921

RESUMO

Despite recent studies depicting the capacity of G protein-coupled receptors (GPCRs) to activate mitogenic signaling pathways more commonly associated with receptor tyrosine kinases (RTKs), little is known regarding the interactive effects of GPCR and RTK activation on cell growth and signal transduction. Such interactions likely mediate the physiologic growth in most cells in vivo as well as the aberrant, non-neoplastic growth that occurs in diseases such as asthma, where disruptions of the local hormonal or inflammatory state can contribute to significant GPCR activation. In this study, we show that numerous inflammatory or contractile agents, including thrombin, histamine, and carbachol, potentiate epidermal growth factor (EGF)-stimulated proliferation of human airway smooth muscle (ASM), thus demonstrating a clear synergy between RTK and GPCR activation. Alterations in promitogenic nuclear signaling were evidenced by additive or synergistic increases in Elk-1 and activator protein-1 activation, and by increases in cyclin D1 expression. Interestingly, GPCR activation did not cause EGF receptor tyrosine phosphorylation nor did it increase EGF-stimulated autophosphorylation. In the presence of EGF, histamine or carbachol did not alter the time-dependent phosphorylation of p42/p44, whereas thrombin was capable of increasing phospho-p42/p44 levels at selected time points in some, but not all, cultures. In contrast to their relative inability to alter EGF receptor-linked p42/p44 activation, thrombin, histamine, and carbachol consistently increased the late phase (> 1 h) activity of p70 S6 kinase. Collectively, these findings suggest that inflammatory and contractile agents that activate GPCRs can significantly modulate RTK-mediated ASM growth through a p70 S6 kinase-dependent, p42/p44-independent mechanism.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Músculo Liso/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Traqueia/efeitos dos fármacos , Carbacol/farmacologia , Células Cultivadas , Ciclina D1/metabolismo , Replicação do DNA/efeitos dos fármacos , Ativação Enzimática , Fator de Crescimento Epidérmico/farmacologia , Histamina/farmacologia , Humanos , Músculo Liso/enzimologia , Fosforilação , Trombina/farmacologia , Traqueia/enzimologia , Traqueia/metabolismo
7.
Am J Physiol ; 277(3): L479-88, 1999 09.
Artigo em Inglês | MEDLINE | ID: mdl-10484455

RESUMO

Asthma is frequently associated with abnormal airway smooth muscle (ASM) growth that may contribute to airway narrowing and hyperresponsiveness to contractile agents. Although numerous hormones and cytokines have been shown to induce human ASM (HASM) proliferation, the cellular and molecular mechanisms underlying HASM hyperplasia are largely unknown. Here we characterize the roles of the mitogen-activated protein kinase (MAPK) superfamily [p42/p44 MAPK, c-Jun amino-terminal kinase/stress-activated protein kinase (JNK/SAPK), and p38] in mediating hormone- and cytokine-induced HASM proliferation. Significant enhancement of [(3)H]thymidine incorporation in HASM cultures was observed only by treatment with agents (epidermal growth factor, platelet-derived growth factor, thrombin, and phorbol 12-myristate 13-acetate) that promoted a strong and sustained activation of p42/p44 MAPK. Significant activation of the JNK/SAPK and p38 pathways was only observed on stimulation with interleukin (IL)-1beta and tumor necrosis factor-alpha, agents that did not appreciably stimulate HASM proliferation. Two different inhibitors of MAPK/extracellular signal-regulated kinase kinase (MEK), PD-98059 and U-0126, inhibited mitogen-induced [3H]thymidine incorporation in a manner consistent with their ability to inhibit p42/p44 activation. Elk-1 and activator protein-1 reporter activation by mitogens was similarly inhibited by inhibition of MEK, suggesting a linkage between p42/p44 activation, transcription factor activation, and HASM proliferation. These findings establish a fundamental role for p42/p44 activation in regulating HASM proliferation and provide insight into species-specific differences observed among studies in ASM mitogenesis.


Assuntos
Proteínas de Ligação a DNA , Regulação da Expressão Gênica , Proteínas Quinases Ativadas por Mitógeno/genética , Família Multigênica/genética , Músculo Liso/fisiologia , Proteínas Serina-Treonina Quinases , Traqueia/fisiologia , Fatores de Transcrição , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Humanos , MAP Quinase Quinase 1 , MAP Quinase Quinase 2 , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Mitose/fisiologia , Músculo Liso/citologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Traqueia/citologia , Fator de Transcrição AP-1/genética , Transcrição Gênica , Proteínas Elk-1 do Domínio ets
8.
Am J Physiol ; 277(1): L65-78, 1999 07.
Artigo em Inglês | MEDLINE | ID: mdl-10409232

RESUMO

Hypertrophy and hyperplasia of airway smooth muscle (ASM) are important pathological features that contribute to airflow obstruction in chronic severe asthma. Despite considerable research effort, the cellular mechanisms that modulate ASM growth remain unknown. Recent evidence suggests that mitogen-induced activation of phosphoinositide (PI)-specific phospholipase C (PLC) and PI-dependent calcium mobilization are neither sufficient nor necessary to stimulate human ASM proliferation. In this study, we identify phosphatidylinositol (PtdIns) 3-kinase as a key regulator of human ASM proliferation. Pretreatment of human ASM with the PtdIns 3-kinase inhibitors wortmannin and LY-294002 significantly reduced thrombin- and epidermal growth factor (EGF)-induced DNA synthesis (IC(50) approximately 10 nM and approximately 3 microM, respectively). In separate experiments, wortmannin and LY-294002 markedly inhibited PtdIns 3-kinase and 70-kDa S6 protein kinase (pp70(S6k)) activation induced by stimulation of human ASM cells with EGF and thrombin but had no effect on EGF- and thrombin-induced p42/p44 mitogen-activated protein kinase (MAPK) activation. The specificity of wortmannin and LY-294002 was further suggested by the demonstrated inability of these compounds to alter thrombin-induced calcium transients, total PI hydrolysis, or basal cAMP levels. Transient expression of constitutively active PtdIns 3-kinase (p110*) activated pp70(S6k), whereas a dominant-negative PtdIns 3-kinase (Deltap85) blocked EGF- and thrombin-stimulated pp70(S6k) activity. Collectively, these data suggest that activation of PtdIns 3-kinase is required for the mitogenic effect of EGF and thrombin in human ASM cells. Further investigation of the role of PtdIns 3-kinase may offer new therapeutic approaches in the treatment of diseases characterized by smooth muscle cell hyperplasia such as asthma and chronic bronchitis.


Assuntos
Mitógenos/farmacologia , Músculo Liso/citologia , Fosfatidilinositol 3-Quinases/fisiologia , Traqueia/citologia , Androstadienos/farmacologia , Cálcio/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Células Cultivadas , Cromonas/farmacologia , AMP Cíclico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Humanos , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositóis/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Quinases S6 Ribossômicas/metabolismo , Trombina/farmacologia , Wortmanina
9.
Am J Physiol ; 276(2): L246-55, 1999 02.
Artigo em Inglês | MEDLINE | ID: mdl-9950886

RESUMO

The epidermal growth factor (EGF)-receptor (EGFR) family includes four homologous transmembrane receptor protein tyrosine kinases, EGFR, ErbB-2, ErbB-3, and ErbB-4. This receptor family plays a pivotal role in regulating cell proliferation, differentiation, and transformation. Ligand-induced activation of these receptors results in formation of homo- and heterodimers, which undergo transphosphorylation and transactivation. The aim of this study was to characterize the role of EGFR family members in signaling EGF-induced human airway smooth muscle (HASM) cell proliferation. Here, we show that EGF stimulates activation of EGFR and transactivation of ErbB-2 in quiescent HASM cells. Phosphatidylinositol (PI) 3-kinase, a critical signaling enzyme that modulates HASM cell growth, is preferentially associated with ErbB-2, and EGF-stimulated transactivation of ErbB-2 induces PI 3-kinase activation. ErbB-3 and ErbB-4 are present in HASM cells; however, EGF has no effect on their activation. Betacellulin, a ligand for EGFR, ErbB-3, and ErbB-4, induced DNA synthesis of HASM cells and stimulated signaling through the activation of EGFR and ErbB-2 but not of ErbB-3 and ErbB-4. Heregulin, a specific ligand for ErbB-3 and ErbB-4, did not effect DNA synthesis and did not activate its specific receptors. These data suggest that EGF imparts signals that involve activation of ErbB-2 and are associated with ErbB-2 PI 3-kinase activation. Despite the mRNA and protein expression of all members of the EGFR family, ligand-stimulated signaling induced activation of EGFR and ErbB-2 but not of ErbB-3 and ErbB-4.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Músculo Liso/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptor ErbB-2/fisiologia , Traqueia/metabolismo , Células Cultivadas , Ativação Enzimática/fisiologia , Receptores ErbB/metabolismo , Receptores ErbB/fisiologia , Humanos , Músculo Liso/citologia , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Receptor ErbB-3 , Receptor ErbB-4 , Traqueia/citologia , Tirosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA