Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Redox Biol ; 6: 565-577, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26496488

RESUMO

Transforming growth factor beta (TGF-ß) is the most potent pro-fibrogenic cytokine and its expression is increased in almost all of fibrotic diseases. Although signaling through Smad pathway is believed to play a central role in TGF-ß's fibrogenesis, emerging evidence indicates that reactive oxygen species (ROS) modulate TGF-ß's signaling through different pathways including Smad pathway. TGF-ß1 increases ROS production and suppresses antioxidant enzymes, leading to a redox imbalance. ROS, in turn, induce/activate TGF-ß1 and mediate many of TGF-ß's fibrogenic effects, forming a vicious cycle (see graphic flow chart on the right). Here, we review the current knowledge on the feed-forward mechanisms between TGF-ß1 and ROS in the development of fibrosis. Therapeutics targeting TGF-ß-induced and ROS-dependent cellular signaling represents a novel approach in the treatment of fibrotic disorders.


Assuntos
Espécies Reativas de Oxigênio/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Animais , Transição Epitelial-Mesenquimal , Fibrose , Humanos , Mitocôndrias/metabolismo , NADPH Oxidases/metabolismo , Oxirredução , Estresse Oxidativo , Inibidor 1 de Ativador de Plasminogênio/metabolismo
2.
J Pharmacol Exp Ther ; 351(1): 87-95, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25047515

RESUMO

Myofibroblasts are effector cells in fibrotic disorders that synthesize and remodel the extracellular matrix (ECM). This study investigated the role of the Src kinase pathway in myofibroblast activation in vitro and fibrogenesis in vivo. The profibrotic cytokine, transforming growth factor ß1 (TGF-ß1), induced rapid activation of Src kinase, which led to myofibroblast differentiation of human lung fibroblasts. The Src kinase inhibitor AZD0530 (saracatinib) blocked TGF-ß1-induced Src kinase activation in a dose-dependent manner. Inhibition of Src kinase significantly reduced α-smooth muscle actin (α-SMA) expression, a marker of myofibroblast differentiation, in TGF-ß1-treated lung fibroblasts. In addition, the induced expression of collagen and fibronectin and three-dimensional collagen gel contraction were also significantly inhibited in AZD0530-treated fibroblasts. The therapeutic efficiency of Src kinase inhibition in vivo was tested in the bleomycin murine lung fibrosis model. Src kinase activation and collagen accumulation were significantly reduced in the lungs of AZD0530-treated mice when compared with controls. Furthermore, the total fibrotic area and expression of α-SMA and ECM proteins were significantly decreased in lungs of AZD0530-treated mice. These results indicate that Src kinase promotes myofibroblast differentiation and activation of lung fibroblasts. Additionally, these studies provide proof-of-concept for targeting the noncanonical TGF-ß signaling pathway involving Src kinase as an effective therapeutic strategy for lung fibrosis.


Assuntos
Benzodioxóis/farmacologia , Diferenciação Celular , Inibidores Enzimáticos/farmacologia , Miofibroblastos/efeitos dos fármacos , Fibrose Pulmonar/tratamento farmacológico , Quinazolinas/farmacologia , Quinases da Família src/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Benzodioxóis/uso terapêutico , Linhagem Celular , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Inibidores Enzimáticos/uso terapêutico , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miofibroblastos/citologia , Miofibroblastos/enzimologia , Quinazolinas/uso terapêutico , Fator de Crescimento Transformador beta/farmacologia , Quinases da Família src/antagonistas & inibidores
3.
J Biol Chem ; 289(26): 18270-8, 2014 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-24831009

RESUMO

Hydrogen peroxide-inducible clone 5 (Hic-5) is a focal adhesion adaptor protein induced by the profibrotic cytokine TGF-ß1. We have demonstrated previously that TGF-ß1 induces myofibroblast differentiation and lung fibrosis by activation of the reactive oxygen species-generating enzyme NADPH oxidase 4 (Nox4). Here we investigated a potential role for Hic-5 in regulating Nox4, myofibroblast differentiation, and senescence. In normal human diploid fibroblasts, TGF-ß1 induces Hic-5 expression in a delayed manner relative to the induction of Nox4 and myofibroblast differentiation. Hic-5 silencing induced constitutive Nox4 expression and enhanced TGF-ß1-inducible Nox4 levels. The induction of constitutive Nox4 protein in Hic-5-silenced cells was independent of transcription and translation and controlled by the ubiquitin-proteasomal system. Hic-5 associates with the ubiquitin ligase Cbl-c and the ubiquitin-binding protein heat shock protein 27 (HSP27). The interaction of these proteins is required for the ubiquitination of Nox4 and for maintaining low basal levels of this reactive oxygen species-generating enzyme. Our model suggests that TGF-ß1-induced Hic-5 functions as a negative feedback mechanism to limit myofibroblast differentiation and senescence by promoting the ubiquitin-proteasomal system-mediated degradation of Nox4. Together, these studies indicate that endogenous Hic-5 suppresses senescence and profibrotic activities of myofibroblasts by down-regulating Nox4 protein expression. Additionally, these are the first studies, to our knowledge, to demonstrate posttranslational regulation of Nox4.


Assuntos
Regulação para Baixo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas com Domínio LIM/metabolismo , NADPH Oxidases/genética , Diferenciação Celular , Fibroblastos/citologia , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Proteínas de Choque Térmico HSP27/genética , Proteínas de Choque Térmico HSP27/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas com Domínio LIM/genética , Miofibroblastos/citologia , Miofibroblastos/enzimologia , Miofibroblastos/metabolismo , NADPH Oxidase 4 , NADPH Oxidases/metabolismo , Proteólise , Fator de Crescimento Transformador beta1/metabolismo , Ubiquitinação
4.
Am J Physiol Gastrointest Liver Physiol ; 306(11): G947-58, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24722904

RESUMO

The intestinal epithelium is subjected to various types of mechanical stress. In this study, we investigated the impact of cyclic stretch on tight junction and adherens junction integrity in Caco-2 cell monolayers. Stretch for 2 h resulted in a dramatic modulation of tight junction protein distribution from a linear organization into wavy structure. Continuation of cyclic stretch for 6 h led to redistribution of tight junction proteins from the intercellular junctions into the intracellular compartment. Disruption of tight junctions was associated with redistribution of adherens junction proteins, E-cadherin and ß-catenin, and dissociation of the actin cytoskeleton at the actomyosin belt. Stretch activates JNK2, c-Src, and myosin light-chain kinase (MLCK). Inhibition of JNK, Src kinase or MLCK activity and knockdown of JNK2 or c-Src attenuated stretch-induced disruption of tight junctions, adherens junctions, and actin cytoskeleton. Paracellular permeability measured by a novel method demonstrated that cyclic stretch increases paracellular permeability by a JNK, Src kinase, and MLCK-dependent mechanism. Stretch increased tyrosine phosphorylation of occludin, ZO-1, E-cadherin, and ß-catenin. Inhibition of JNK or Src kinase attenuated stretch-induced occludin phosphorylation. Immunofluorescence localization indicated that phospho-MLC colocalizes with the vesicle-like actin structure at the actomyosin belt in stretched cells. On the other hand, phospho-c-Src colocalizes with the actin at the apical region of cells. This study demonstrates that cyclic stretch disrupts tight junctions and adherens junctions by a JNK2, c-Src, and MLCK-dependent mechanism.


Assuntos
Ativação Enzimática/fisiologia , Genes src/fisiologia , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Junções Íntimas/fisiologia , Actinas/fisiologia , Junções Aderentes/fisiologia , Antracenos , Células CACO-2 , Humanos , Mecânica , Quinase de Cadeia Leve de Miosina/genética , Periodicidade , Fosforilação , Pirimidinas , Tirosina/análogos & derivados
5.
Am J Physiol Lung Cell Mol Physiol ; 301(3): L275-84, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21642449

RESUMO

Airway smooth muscle phenotype may be modulated in response to external stimuli under physiological and pathophysiological conditions. The effect of mechanical forces on airway smooth muscle phenotype were evaluated in vitro by suspending weights of 0.5 or 1 g from the ends of canine tracheal smooth muscle tissues, incubating the weighted tissues for 6 h, and then measuring the expression of the phenotypic marker protein, smooth muscle myosin heavy chain (SmMHC). Incubation of the tissues at a high load significantly increased expression of SmMHC compared with incubation at low load. Incubation of the tissues at a high load also decreased activation of PKB/Akt, as indicated by its phosphorylation at Ser 473. Inhibition of Akt or phosphatidylinositol-3,4,5 triphosphate-kinase increased SmMHC expression in tissues at low load but did not affect SmMHC expression at high load. IL-13 induced a significant increase in Akt activation and suppressed the expression of SmMHC protein at both low and high loads. The role of integrin signaling in mechanotransduction was evaluated by expressing a PINCH (LIM1-2) fragment in the muscle tissues that prevents the membrane localization of the integrin-binding IPP complex (ILK/PINCH/α-parvin), and also by expressing an inactive integrin-linked kinase mutant (ILK S343A) that inhibits endogenous ILK activity. Both mutants inhibited Akt activation and increased expression of SmMHC protein at low load but had no effect at high load. These results suggest that mechanical stress and IL-13 both act through an integrin-mediated signaling pathway to oppositely regulate the expression of phenotypic marker proteins in intact airway smooth muscle tissues. The stimulatory effects of mechanical stress on contractile protein expression oppose the suppression of contractile protein expression mediated by IL-13; thus the imposition of mechanical strain may inhibit changes in airway smooth muscle phenotype induced by inflammatory mediators.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Interleucina-13/farmacologia , Proteínas dos Microfilamentos/metabolismo , Complexos Multiproteicos/metabolismo , Cadeias Pesadas de Miosina/biossíntese , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Miosinas de Músculo Liso/biossíntese , Estresse Mecânico , Traqueia/fisiologia , Animais , Cães , Mecanotransdução Celular , Contração Muscular/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Serina-Treonina Quinases/genética , Miosinas de Músculo Liso/metabolismo
6.
J Biol Chem ; 285(7): 4511-9, 2010 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-20018857

RESUMO

Epithelial cell migration during wound healing requires coordinated signaling pathways that direct polarization of the leading and trailing ends of the cells, cytoskeletal organization, and remodeling of focal adhesions. These inherently mechanical processes are disrupted by cyclic stretch (CS), but the specific signaling molecules involved in this disruption are not well understood. In this study, we demonstrate that inhibition of phosphatidylinositol 3-kinase (PI3K) or expression of a dominant-negative form of PI3K caused inhibition of airway epithelial cell wound closure. CS caused a sustained decrease in activation of PI3K and inhibited wound healing. Expression of constitutively active PI3K stimulated translocation of Tiam1 to the membrane, increased Rac1 activity, and increased wound healing of airway epithelial cells. Increased Rac1 activity resulted in increased phosphorylation of JNK1. PI3K activation was not regulated by association with focal adhesion kinase. Restoration of efficient cell migration during CS required coexpression of constitutively active PI3K, focal adhesion kinase, and JIP3.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Estresse Mecânico , Animais , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Cromonas/farmacologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Immunoblotting , Imunoprecipitação , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Ratos , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
7.
Am J Physiol Lung Cell Mol Physiol ; 297(3): L520-9, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19574423

RESUMO

JNK is a nonreceptor kinase involved in the early events that signal cell migration after injury. However, the linkage to early signals required to initiate the migration response to JNK has not been defined in airway epithelial cells, which exist in an environment subjected to cyclic mechanical strain (MS). The present studies demonstrate that the JNK/stress-activated protein kinase-associated protein 1 (JSAP1; also termed JNK-interacting protein 3, JIP3), a scaffold factor for MAPK cascades that links JNK activation to focal adhesion kinase (FAK), are both associated and activated following mechanical injury in 16HBE14o- human airway epithelial cells and that both FAK and JIP3 phosphorylation seen after injury are decreased in cells subjected to cyclic MS. Overexpression of either wild-type (WT)-FAK or WT-JIP3 enhanced phosphorylation and kinase activation of JNK and reduced the inhibitory effect of cyclic MS. These results suggest that cyclic MS impairs signaling of cell migration after injury via a pathway that involves FAK-JIP3-JNK.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Movimento Celular , Células Epiteliais/enzimologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Pulmão/citologia , Proteínas do Tecido Nervoso/metabolismo , Estresse Mecânico , Animais , Linhagem Celular , Ativação Enzimática , Células Epiteliais/citologia , Humanos , Proteína Quinase 8 Ativada por Mitógeno , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Modelos Biológicos , Fosforilação , Ligação Proteica , Ratos , Cicatrização
8.
Am J Physiol Lung Cell Mol Physiol ; 295(5): L958-65, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18805958

RESUMO

Mechanical ventilation can overdistend the lungs or generate shear forces in them during repetitive opening/closing, contributing to lung injury and inflammation in patients with acute respiratory distress syndrome (ARDS). Repair of the injured lung epithelium is important for restoring normal barrier and lung function. In the current study, we investigated the effects of cyclic mechanical strain (CS), constant distention strain (CD), and simulated positive end-expiratory pressure (PEEP) on activation of Rac1 and wound closure of rat primary alveolar type 2 (AT2) cells. Cyclic stretch inhibited the migration of wounded AT2 cells in a dose-dependent manner with no inhibition occurring with 5% CS, but significant inhibition with 10% and 15% CS. PEEP conditions were investigated by stretching AT2 cells to 15% maximum strain (at a frequency of 10 cycles/min) with relaxation to 10% strain. AT2 cells were also exposed to 20% CD. All three types of mechanical strain inhibited wound closure of AT2 cells compared with static controls. Since lamellipodial extensions in migrating cells at the wound edge were significantly smaller in stretched cells, we measured Rac1 activity and found it to be decreased in stretched cells. We also demonstrate that Tiam1, a Rac1-specific guanine nucleotide exchange factor, was expressed mainly in the cytosol of AT2 cells exposed to mechanical strain compared with membrane localization in static cells. Downregulation of Tiam1 with 100 microM NSC-23766 inhibited activation of Rac1 and migration of AT2 cells, suggesting its involvement in repair mechanisms of AT2 cells subjected to mechanical strain.


Assuntos
Movimento Celular , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas de Neoplasias/metabolismo , Alvéolos Pulmonares/citologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Membrana Celular/enzimologia , Citosol/enzimologia , Regulação para Baixo , Ativação Enzimática , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Humanos , Masculino , Proteínas de Neoplasias/antagonistas & inibidores , Transporte Proteico , Pseudópodes/metabolismo , Ratos , Ratos Sprague-Dawley , Estresse Mecânico , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T
9.
Am J Physiol Lung Cell Mol Physiol ; 295(1): L54-60, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18487359

RESUMO

Restoration of lung homeostasis following injury requires efficient wound healing by the epithelium. The mechanisms of lung epithelial wound healing include cell spreading and migration into the wounded area and later cell proliferation. We hypothesized that mechanical properties of cells vary near the wound edge, and this may provide cues to direct cell migration. To investigate this hypothesis, we measured variations in the stiffness of migrating human bronchial epithelial cells (16HBE cells) approximately 2 h after applying a scratch wound. We used atomic force microscopy (AFM) in contact mode to measure the cell stiffness in 1.5-microm square regions at different locations relative to the wound edge. In regions far from the wound edge (>2.75 mm), there was substantial variation in the elastic modulus in specific cellular regions, but the median values measured from multiple fields were consistently lower than 5 kPa. At the wound edge, cell stiffness was significantly lower within the first 5 microm but increased significantly between 10 and 15 microm before decreasing again below the median values away from the wound edge. When cells were infected with an adenovirus expressing a dominant negative form of RhoA, cell stiffness was significantly decreased compared with cells infected with a control adenovirus. In addition, expression of dominant negative RhoA abrogated the peak increase in stiffness near the wound edge. These results suggest that cells near the wound edge undergo localized changes in cellular stiffness that may provide signals for cell spreading and migration.


Assuntos
Movimento Celular , Células Epiteliais/patologia , Microscopia de Força Atômica , Mucosa Respiratória/patologia , Cicatrização , Ferimentos e Lesões/patologia , Adenoviridae , Linhagem Celular , Movimento Celular/genética , Elasticidade , Células Epiteliais/enzimologia , Humanos , Mutação , Mucosa Respiratória/enzimologia , Cicatrização/genética , Ferimentos e Lesões/enzimologia , Ferimentos e Lesões/genética , Proteína rhoA de Ligação ao GTP/biossíntese , Proteína rhoA de Ligação ao GTP/genética
10.
Am J Physiol Lung Cell Mol Physiol ; 293(3): L769-78, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17601798

RESUMO

Patients with acute respiratory distress syndrome undergoing mechanical ventilation may be exposed to both high levels of stretch and high levels of oxygen. We hypothesized that the combination of high stretch and hyperoxia promotes loss of epithelial adhesion and impairs epithelial repair mechanisms necessary for restoration of barrier function. We utilized a model of high tidal volume mechanical ventilation (25 ml/kg) with hyperoxia (50% O(2)) in rats to investigate alveolar type II (AT2) cell adhesion and focal adhesion signaling. AT2 cells isolated from rats exposed to hyperoxia and high tidal volume mechanical ventilation (MVHO) exhibited significantly decreased cell adhesion and reduction in phosphotyrosyl levels of focal adhesion kinase (FAK) and paxillin compared with control rats, rats exposed to hyperoxia without ventilation (HO), or rats ventilated with normoxia (MV). MV alone increased phosphorylation of p130(Cas). RhoA activation was increased by MV, HO, and the combination of MV and HO. Treatment of MVHO cells with keratinocyte growth factor (KGF) for 1 h upon isolation reduced RhoA activity and restored attachment to control levels. Attachment and migration of control AT2 cells was significantly decreased by constitutively active RhoA or a kinase inactive form of FAK (FRNK), whereas expression of dominant negative RhoA in cells from MVHO-treated rats restored cell adhesion. Mechanical ventilation with hyperoxia promotes changes in focal adhesion proteins and RhoA in AT2 cells that may be deleterious for cell adhesion and migration.


Assuntos
Hiperóxia/patologia , Alvéolos Pulmonares/citologia , Respiração Artificial , Animais , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Separação Celular , Proteína Substrato Associada a Crk/metabolismo , Ativação Enzimática/efeitos dos fármacos , Fator 7 de Crescimento de Fibroblastos/farmacologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Genes Dominantes , Humanos , Hiperóxia/induzido quimicamente , Paxilina/metabolismo , Fosforilação/efeitos dos fármacos , Fosfotirosina/metabolismo , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/enzimologia , Ratos , Ratos Sprague-Dawley , Volume de Ventilação Pulmonar/efeitos dos fármacos , Proteína rhoA de Ligação ao GTP/metabolismo
11.
Am J Physiol Lung Cell Mol Physiol ; 289(5): L834-41, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15964900

RESUMO

Overdistention of lung tissue during mechanical ventilation may be one of the factors that initiates ventilator-induced lung injury (VILI). We hypothesized that cyclic mechanical stretch (CMS) of the lung epithelium is involved in the early events of VILI through the production of reactive oxygen species (ROS). Cultures of an immortalized human airway epithelial cell line (16HBE), a human alveolar type II cell line (A549), and primary cultures of rat alveolar type II cells were cyclically stretched, and the production of superoxide (O2-) was measured by dihydroethidium fluorescence. CMS stimulated increased production of O2- after 2 h in each type of cell. 16HBE cells exhibited no significant stimulation of ROS before 2 h of CMS (20% strain, 30 cycles/min), and ROS production returned to control levels after 24 h. Oxidation of glutathione (GSH), a cellular antioxidant, increased with CMS as measured by a decrease in the ratio of the reduced GSH level to the oxidized GSH level. Strain levels of 10% did not increase O2- production in 16HBE cells, whereas 15, 20, and 30% significantly increased generation of O2-. Rotenone, a mitochondrial complex I inhibitor, partially abrogated the stretch-induced generation of O2- after 2 h CMS in 16HBE cells. NADPH oxidase activity was increased after 2 h of CMS, contributing to the production of O2-. Increased ROS production in lung epithelial cells in response to elevated stretch may contribute to the onset of VILI.


Assuntos
Pulmão/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Células Epiteliais/metabolismo , Glutationa/metabolismo , Humanos , Pulmão/citologia , Mecanotransdução Celular , Mitocôndrias/metabolismo , NADPH Oxidases/metabolismo , Oxirredução , Ratos , Respiração Artificial/efeitos adversos , Síndrome do Desconforto Respiratório/etiologia , Síndrome do Desconforto Respiratório/metabolismo , Superóxidos/metabolismo
12.
Am J Physiol Lung Cell Mol Physiol ; 287(6): L1134-44, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15298851

RESUMO

Repair of the airway epithelium after injury is critical for restoring normal lung. The reepithelialization process involves spreading and migration followed later by cell proliferation. Rho-GTPases are key components of the wound healing process in many different types of tissues, but the specific roles for RhoA and Rac1 vary and have not been identified in lung epithelial cells. We investigated whether RhoA and Rac1 regulate wound closure of bronchial epithelial cells. RhoA and Rac1 proteins were efficiently expressed in a cell line of human bronchial epithelial cells (16HBE) by adenovirus-based gene transfer. We found that both constitutively active RhoA and dominant negative RhoA inhibited wound healing, suggesting that both activation and inhibition of RhoA interfere with normal wound healing. Overexpression of wild-type Rac1 induced upregulation of RhoA, disrupted intercellular junctions, and inhibited wound closure. Dominant negative Rac1 also inhibited wound closure. Inhibition of the downstream effector of RhoA, Rho-kinase, with Y-27632 suppressed actin stress fibers and focal adhesion formation, increased Rac1 activity, and stimulated wound closure. The activity of both RhoA and Rac1 are influenced by the polymerization state of microtubules, and cell migration involves coordinated action of actin and microtubules. Microtubule depolymerization upon nocodazole treatment led to an increase in focal adhesions and decreased wound closure. We conclude that coordination of both RhoA and Rac1 activity contributes to bronchial epithelial wound repair mechanisms in vitro, that inhibition of Rho-kinase accelerates wound closure, and that efficient repair involves intact microtubules.


Assuntos
Mucosa Respiratória/fisiologia , Cicatrização/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Amidas/farmacologia , Brônquios , Linhagem Celular , Humanos , Cinética , Piridinas/farmacologia , Cicatrização/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...