Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nat Med ; 10(2): 193-6, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14716307

RESUMO

Mucus hypersecretion is a crucial feature of pulmonary diseases such as asthma, chronic bronchitis and cystic fibrosis. Despite much research, there is still no effective therapy for this condition. Recently, we showed that the myristoylated, alanine-rich C-kinase substrate (MARCKS) protein is required for mucus secretion by human bronchial epithelial cells in culture. Having synthesized a peptide corresponding to the N-terminal domain of MARCKS, we now show that the intratracheal instillation of this peptide blocks mucus hypersecretion in a mouse model of asthma. A missense peptide with the same amino acid composition has no effect. Based on quantitative histochemical analysis of the mouse airways, the peptide seems to act by blocking mucus release from goblet cells, possibly by inhibiting the attachment of MARCKS to membranes of intracellular mucin granules. These results support a pivotal role for MARCKS protein, specifically its N-terminal region, in modulating this secretory process in mammalian airways. Intratracheal administration of this MARCKS-related peptide could therapeutically reduce mucus secretion in the airways of human patients with asthma, chronic bronchitis and cystic fibrosis.


Assuntos
Asma/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Muco/metabolismo , Fragmentos de Peptídeos/farmacologia , Peptídeos/farmacologia , Proteínas/metabolismo , Animais , Brônquios/citologia , Brônquios/metabolismo , Brônquios/patologia , Testes de Provocação Brônquica , Grânulos Citoplasmáticos/metabolismo , Grânulos Citoplasmáticos/ultraestrutura , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mucinas/metabolismo , Substrato Quinase C Rico em Alanina Miristoilada , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/química , Peptídeos/administração & dosagem , Peptídeos/química , Proteínas/química , Proteínas/genética , Mucosa Respiratória/citologia , Mucosa Respiratória/metabolismo
2.
Am J Physiol Lung Cell Mol Physiol ; 299(5): L681-93, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20729386

RESUMO

IL-4 and IL-13 elicit several important responses in airway epithelium including chemokine secretion and mucous secretion that may contribute to airway inflammation, cell migration, and differentiation. These cytokines have overlapping but not identical effector profiles likely due to shared subunits in their receptor complexes. These receptors are variably described in epithelial cells, and the relative expression, localization, and function of these receptors in differentiated and repairing epithelial cells are not clear. We examined IL-4/IL-13 receptor expression and localization in primary airway epithelial cells collected from normal human lungs and grown under conditions yielding both undifferentiated and differentiated cells inclusive of basal, goblet, and ciliated cell phenotypes. Gene expression of the IL-4Rα, IL-2Rγc, IL-13Rα1, and IL-13Rα2 receptor subunits increased with differentiation, but different patterns of localization and protein abundance were seen for each subunit based on both differentiation and the cell subtypes present. Increased expression of receptor subunits observed in more differentiated cells was associated with more substantial functional responses to IL-4 stimulation including increased eotaxin-3 expression and accelerated migration after injury. We demonstrate substantial differences in IL-4/IL-13 receptor subunit expression and responsiveness to IL-4 based on the extent of airway epithelial cell differentiation and suggest that these differences may have functional consequences in airway inflammation.


Assuntos
Diferenciação Celular/fisiologia , Células Epiteliais/fisiologia , Receptores de Interleucina-13/metabolismo , Receptores de Interleucina-4/metabolismo , Mucosa Respiratória/citologia , Animais , Movimento Celular , Células Cultivadas , Quimiocina CCL26 , Quimiocinas CC/genética , Quimiocinas CC/metabolismo , Células Epiteliais/citologia , Humanos , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Receptores de Interleucina-13/genética , Receptores de Interleucina-4/genética , Mucosa Respiratória/fisiologia , Estresse Mecânico
3.
J Toxicol Environ Health A ; 73(8): 565-80, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20391136

RESUMO

Exposure to traffic-related ambient air pollution, such as diesel exhaust particles (DEP), is associated with adverse health outcomes, especially in individuals with preexisting inflammatory respiratory diseases. Using an analogous novel in vitro system to model both the healthy and inflamed lung, the susceptibility of epithelial cells exposed to DEP of varying organic carbon content was studied. Murine LA-4 alveolar type II-like epithelial cells, as well as primary murine tracheal epithelial cells (MTE), were treated with exogenous cytokines (tumor necrosis factor [TNF] alpha + interleukin [IL]-1 beta + interferon [IFN] gamma) to model a mild inflammatory state. Epithelial cells were subsequently exposed to DEP of varying organic carbon content, and the resultant cytotoxic, cytoprotective, or antioxidant cell responses were inferred by changes in lactate dehydrogenase (LDH) release, heme oxygenase-1 (HO-1) expression, or glutathione levels, respectively. Data showed that exposure of healthy LA-4 cells to organic carbon-rich DEP (25 microg/cm(2); 24 h) induced adaptive cytoprotective/antioxidant responses with no apparent cell injury. In contrast, exposure of inflamed LA-4 cells resulted in oxidative stress culminating in significant cytotoxicity. Exposure of healthy MTE cells to organic carbon-rich DEP (20 microg/cm(2); 24 h) was seemingly without effect, whereas exposure of inflamed MTE cells resulted in increased epithelial solute permeability. Thus, surface lung epithelial cells stressed by a state of inflammation and then exposed to organic carbon-rich DEP appear unable to respond to the additional oxidative stress, resulting in epithelial barrier dysfunction and injury. Adverse health outcomes associated with exposure to traffic-related air pollutants, like DEP, in patients with preexisting inflammatory respiratory diseases may be due, in part, to similar mechanisms.


Assuntos
Poluentes Atmosféricos/toxicidade , Carbono/toxicidade , Material Particulado/toxicidade , Alvéolos Pulmonares/efeitos dos fármacos , Mucosa Respiratória/efeitos dos fármacos , Emissões de Veículos/toxicidade , Actinas/metabolismo , Poluentes Atmosféricos/química , Animais , Células Cultivadas , Glutationa/metabolismo , Heme Oxigenase-1/metabolismo , L-Lactato Desidrogenase/metabolismo , Camundongos , Material Particulado/química , Mucosa Respiratória/citologia
4.
Am J Physiol Lung Cell Mol Physiol ; 297(1): L164-73, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19447894

RESUMO

Migration of airway epithelial cells (AEC) is an integral component of airway mucosal repair after injury. The inflammatory cytokine IL-4, abundant in chronic inflammatory airways diseases such as asthma, stimulates overproduction of mucins and secretion of chemokines from AEC; these actions enhance persistent airway inflammation. The effect of IL-4 on AEC migration and repair after injury, however, is not known. We examined migration in primary human AEC differentiated in air-liquid interface culture for 3 wk. Wounds were created by mechanical abrasion and followed to closure using digital microscopy. Concurrent treatment with IL-4 up to 10 ng/ml accelerated migration significantly in fully differentiated AEC. As expected, IL-4 treatment induced phosphorylation of the IL-4 receptor-associated protein STAT (signal transducer and activator of transcription)6, a transcription factor known to mediate several IL-4-induced AEC responses. Expressing a dominant negative STAT6 cDNA delivered by lentivirus infection, however, failed to block IL-4-stimulated migration. In contrast, decreasing expression of either insulin receptor substrate (IRS)-1 or IRS-2 using a silencing hairpin RNA blocked IL-4-stimulated AEC migration completely. These data demonstrate that IL-4 can accelerate migration of differentiated AEC after injury. This reparative response does not require STAT6 activation, but rather requires IRS-1 and/or IRS-2.


Assuntos
Movimento Celular/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Proteínas Substratos do Receptor de Insulina/metabolismo , Interleucina-4/farmacologia , Pulmão/citologia , Ar , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , DNA Complementar/genética , Células Epiteliais/metabolismo , Genes Dominantes , Humanos , Pulmão/patologia , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Fator de Transcrição STAT6/metabolismo
5.
Respir Res ; 8: 51, 2007 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-17620132

RESUMO

BACKGROUND: The pleiotrophic cytokine interleukin (IL)-13 features prominently in allergic and inflammatory diseases. In allergic asthma, IL-13 is well established as an inducer of airway inflammation and tissue remodeling. We demonstrated previously that IL-13 induces release of transforming growth factor-alpha (TGFalpha) from human bronchial epithelial cells, with proliferation of these cells mediated by the autocrine/paracrine action of this growth factor. TGFalpha exists as an integral membrane protein and requires proteolytic processing to its mature form, with a disintegrin and metalloproteinase (ADAM)17 responsible for this processing in a variety of tissues. METHODS: In this study, normal human bronchial epithelial (NHBE) cells grown in air/liquid interface (ALI) culture were used to examine the mechanisms whereby IL-13 induces release of TGFalpha and cellular proliferation. Inhibitors and antisense RNA were used to examine the role of ADAM17 in these processes, while IL-13-induced changes in the intracellular expression of TGFalpha and ADAM17 were visualized by confocal microscopy. RESULTS: IL-13 was found to induce proliferation of NHBE cells, and release of TGFalpha, in an ADAM17-dependent manner; however, this IL-13-induced proliferation did not appear to result solely from ADAM17 activation. Rather, IL-13 induced a change in the location of TGFalpha expression from intracellular to apical regions of the NHBE cells. The apical region was also found to be a site of significant ADAM17 expression, even prior to IL-13 stimulation. CONCLUSION: Results from this study indicate that ADAM17 mediates IL-13-induced proliferation and TGFalpha shedding in NHBE cells. Furthermore, they provide the first example wherein a cytokine (IL-13) induces a change in the intracellular expression pattern of a growth factor, apparently inducing redistribution of intracellular stores of TGFalpha to the apical region of NHBE cells where expression of ADAM17 is prominent. Thus, IL-13-induced, ADAM17-mediated release of TGFalpha, and subsequent epithelial cell proliferation, could contribute to the epithelial hypertrophy, as well as other features, associated with airway remodeling in allergic asthma.


Assuntos
Proteínas ADAM/metabolismo , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/fisiologia , Interleucina-13/farmacologia , Fator de Crescimento Transformador alfa/metabolismo , Análise de Variância , Western Blotting , Brônquios/citologia , Brônquios/efeitos dos fármacos , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Receptores ErbB/antagonistas & inibidores , Humanos , Imunoprecipitação , Microscopia Confocal , Probabilidade , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sensibilidade e Especificidade , Transdução de Sinais/efeitos dos fármacos
6.
Respir Res ; 7: 35, 2006 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-16504136

RESUMO

BACKGROUND: Mucus overproduction is a characteristic of inflammatory pulmonary diseases including asthma, chronic bronchitis, and cystic fibrosis. Expression of two mucin genes, MUC2 and MUC5AC, and their protein products (mucins), is modulated in certain disease states. Understanding the signaling mechanisms that regulate the production and secretion of these major mucus components may contribute significantly to development of effective therapies to modify their expression in inflamed airways. METHODS: To study the differential expression of Muc2 and Muc5ac, a novel monoclonal antibody recognizing guinea pig Muc2 and a commercially-available antibody against human MUC5AC were optimized for recognition of specific guinea pig mucins by enzyme-linked immunosorbent assay (ELISA), Western blot, and immunohistochemistry (IHC). These antibodies were then used to analyze expression of Muc2 and another mucin subtype (likely Muc5ac) in guinea pig tracheal epithelial (GPTE) cells stimulated with a mixture of pro-inflammatory cytokines [tumor necrosis factor-alpha (TNF-alpha), interleukin 1beta (IL-1beta), and interferon- gamma (IFN-gamma)]. RESULTS: The anti-Muc2 (C4) and anti-MUC5AC (45M1) monoclonal antibodies specifically recognized proteins located in Muc2-dominant small intestinal and Muc5ac-dominant stomach mucosae, respectively, in both Western and ELISA experimental protocols. IHC protocols confirmed that C4 recognizes murine small intestine mucosal proteins while 45M1 does not react. C4 and 45M1 also stained specific epithelial cells in guinea pig lung sections. In the resting state, Muc2 was recognized as a highly expressed intracellular mucin in GPTE cells in vitro. Following cytokine exposure, secretion of Muc2, but not the mucin recognized by the 45M1 antibody (likely Muc5ac), was increased from the GPTE cells, with a concomitant increase in intracellular expression of both mucins. CONCLUSION: Given the tissue specificity in IHC and the differential hybridization to high molecular weight proteins by Western blot, we conclude that the antibodies used in this study can recognize specific mucin subtypes in guinea pig airway epithelium and in proteins from GPTE cells. In addition, Muc2 is highly expressed constitutively, modulated by inflammation, and secreted differentially (as compared to Muc5ac) in GPTE cells. This finding contrasts with expression patterns in the airway epithelium of a variety of mammalian species in which only Muc5ac predominates.


Assuntos
Células Epiteliais/metabolismo , Mucinas/metabolismo , Traqueia/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Células Cultivadas , Citocinas , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica , Cobaias , Imuno-Histoquímica , Mucina-5AC , Mucina-2 , Mucinas/imunologia , RNA Mensageiro/análise , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo
7.
FASEB J ; 19(10): 1374-6, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15923396

RESUMO

In cystic fibrosis and chronic bronchitis, airways are chronically injured by exposure to neutrophil elastase (NE). We sought to identify factors required for epithelial repair following NE exposure. Normal human bronchial epithelial cells were treated with NE (50 nM, 22 h) or control vehicle. Following NE treatment, we found a marked and sustained decrease in epithelial proliferation as detected by Ki67 immunostaining. 3H-thymidine incorporation was also initially depressed but increased over 72 h in NE-treated cells, which suggests that DNA synthesis constitutes an early repair process following NE exposure. We hypothesized that ErbB2 receptor tyrosine kinase, a regulator of cancer cell proliferation, was required for epithelial DNA synthesis following NE exposure. Immediately following NE treatment, by flow cytometry analysis, we found a decrease in ErbB2 surface expression. Protein levels of the full-length 185 kD ErbB2 receptor significantly decreased following NE treatment and smaller ErbB2-positive bands, ranging in size from 23 to 40 kD, appeared, which suggests that NE caused ErbB2 degradation. By real-time RT-PCR analysis, we found no change in ErbB2 mRNA expression following NE treatment, which suggests that changes in ErbB2 protein levels were regulated at the post-translational level. Following NE treatment, full-length 185 kD ErbB2 levels increased to pretreatment levels, correlating with the increase in thymidine incorporation during the same time period. Importantly, inhibition of ErbB2 activity with AG825 (5 microM) or Herceptin (3.1 microM), an ErbB2-neutralizing antibody, blocked thymidine incorporation only in NE-treated cells. These results suggest ErbB2 is a critical factor for epithelial recovery following NE exposure.


Assuntos
Brônquios/efeitos dos fármacos , Elastase de Leucócito/toxicidade , Receptor ErbB-2/fisiologia , Benzotiazóis , Brônquios/citologia , Brônquios/fisiologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , DNA/biossíntese , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Receptores ErbB/metabolismo , Humanos , Receptor ErbB-2/antagonistas & inibidores , Tirfostinas/farmacologia
8.
J Vet Intern Med ; 20(1): 47-51, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16496922

RESUMO

Diabetes mellitus (DM) is a common endocrinopathy of cats and humans. Although few studies have examined the effects of DM on the pulmonary system, changes in pulmonary function and immunology in humans with type I and II diabetes, and pulmonary lesions in a murine diabetic model have been documented. Our objective was to determine whether pulmonary lesions occurred in cats with DM. Medical records and necropsy evaluations of 42 cats with DM were compared with those of 45 age-matched, nondiabetic cats for the presence of clinical evidence of respiratory disease and pulmonary histopathological findings at the time of necropsy. No statistical difference was noted in the presence of clinical evidence of respiratory disease between cats with diabetes and control cats. Nevertheless, there was a significant association between the presence of abnormal pulmonary histopathology and DM (P = .018, odds ratio = 3 inclusive of all cats; P = .005, odds ratio = 5 when non-DM cats with overt clinical evidence of respiratory disease were excluded). Pulmonary abnormalities detected by histopathological examination in cats with diabetes included congestion and edema, histiocytosis, pneumonia, smooth muscle hypertrophy, fibrosis, mineralization, neoplasia, and type II pneumocyte hyperplasia. The observed association between DM and pulmonary lesions in cats, independent of clinical evidence of respiratory disease, emphasizes the need for careful assessment of the respiratory tract in sick cats with diabetes.


Assuntos
Doenças do Gato/patologia , Complicações do Diabetes/veterinária , Pneumopatias/veterinária , Animais , Estudos de Casos e Controles , Gatos , Complicações do Diabetes/patologia , Feminino , Pneumopatias/complicações , Pneumopatias/patologia , Masculino
9.
In Vitro Cell Dev Biol Anim ; 41(8-9): 258-63, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16409111

RESUMO

Anurans are important animal models for studying the effects of anthropogenic chemical contamination of the environment. Two-compartment Teflon flow-through diffusion cells can be used to study percutaneous absorption of xenobiotics across harvested skin. However, such an approach currently necessitates that skin be harvested just before experimentation, a requirement that calls for the continuous growth and housing of living animals. The ability to preserve and store skin would allow more efficient use of animals and more flexibility in experimental design. To this end, we examined the viability of harvested anuran skin stored under various protocols consistent with current practices of mammalian skin preservation. Skin from the American bullfrog maintained 80-85% viability after 28 d, whereas viability of skin from the marine toad was only maintained for 7-10 d.


Assuntos
Anuros , Criopreservação/métodos , Glicerol/metabolismo , Absorção Cutânea/fisiologia , Pele/citologia , Preservação de Tecido/métodos , Animais
10.
In Vitro Cell Dev Biol Anim ; 41(7): 217-24, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16223336

RESUMO

Mucous cells of the airway epithelium play a crucial role in the pathogenesis of human inflammatory airway diseases. Therefore, it is of importance to complement in vivo studies that use murine models of allergic asthma with in vitro mechanistic studies that use murine airway epithelial cells, including mucus-containing cells. In this study, we report the development and characterization of an in vitro culture system for primary murine tracheal epithelial (MTE) cells comprising ciliated cells and a substantial number of mucous cells. The increase in mucous cell number over that observed in the native murine airway, or in previously described murine cultures, creates a culture intermediate between the in vivo murine airway epithelium and in vitro cultures of human airway epithelial cells. To establish the usefulness of this culture system for the study of epithelial effects during inflammatory airway diseases, the cells were exposed to interleukin (IL)-13, a central inflammatory mediator in allergic asthma. The IL-13 induced two characteristic epithelial effects, proliferation and modulation of MUC5AC gene expression. There was a concentration dependence of these events, wherein high concentrations of IL-13 (10 ng/ml) induced proliferation, whereas lower concentrations (1 ng/ml) increased MUC5AC mRNA (where mRNA is messenger RNA). Interestingly, these effects occurred in an inverse manner, with the high concentration of IL-13 also provoking a significant decrease in MUC5AC gene expression. Thus, MTE cells cultured in this manner may provide an important link between experimental findings from animal models of allergic asthma and their application to human disease.


Assuntos
Asma/imunologia , Asma/fisiopatologia , Técnicas de Cultura de Células , Células Epiteliais , Interleucina-13/farmacologia , Mucosa Respiratória/citologia , Animais , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Camundongos , Mucina-5AC , Mucinas/metabolismo , Mucosa Respiratória/imunologia , Traqueia/anatomia & histologia
11.
Free Radic Biol Med ; 35(9): 1158-67, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14572618

RESUMO

We demonstrate that two different cell-permeable antioxidants, pyrrolidine dithiocarbamate (PDTC) and dimethylthiourea (DMTU), inhibit TNFalpha-induced ICAM-1 surface and gene expression in primary cultures of differentiated normal human bronchial epithelial (NHBE) cells. In addition, TNFalpha stimulates binding of nuclear proteins to the nuclear factor kappa beta (NFkappaB) and the CAAT/enhancer binding protein (C/EBP) consensus sites in the ICAM-1 promoter in these cells. Because these transcription factors have been suggested to be oxidant-sensitive and important in ICAM-1 expression, the potential involvement of reactive oxygen species (ROS) in the response to TNFalpha was investigated. Interestingly, neither PDTC nor DMTU altered binding of NFkappaB complexes. In contrast, either the proteasome inhibitor carbobenzoxy-L-leucy-L-leucy-L-leucinal (MG 132) or the IkappaBalpha inhibitor BAY 11-7082 ablated TNFalpha-induced ICAM-1 gene expression and MG132 inhibited TNFalpha-induced NFkappaB complexes. Surprisingly, either PDTC or DMTU inhibited the binding of TNFalpha-enhanced C/EBP complexes to the consensus site directly adjacent to the NFkappaB site. These results suggest that although TNFalpha enhances binding of C/EBP and NFkappaB complexes in NHBE cells, C/EBP binding seems to involve an oxidant-dependent mechanism, whereas activation of NFkappaB complexes utilizes the ubiquitin-proteasome pathway, a mechanism that seems to be unaltered by the presence of antioxidants. Because interference with either signaling pathway abrogates TNFalpha-induced ICAM-1 expression, activation of both complexes seems to be involved in this response to TNFalpha, but this activation occurs via different intracellular pathways.


Assuntos
Brônquios/citologia , Células Epiteliais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Molécula 1 de Adesão Intercelular/genética , Oxidantes/metabolismo , Tioureia/análogos & derivados , Fatores de Transcrição/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Antioxidantes/farmacologia , Antioxidantes/toxicidade , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Linhagem Celular , Sequência Consenso/genética , Cisteína Endopeptidases/metabolismo , Células Epiteliais/metabolismo , Humanos , Leupeptinas/farmacologia , Complexos Multienzimáticos/antagonistas & inibidores , Complexos Multienzimáticos/metabolismo , NF-kappa B/metabolismo , Regiões Promotoras Genéticas/genética , Complexo de Endopeptidases do Proteassoma , Pirrolidinas/farmacologia , Pirrolidinas/toxicidade , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Elementos de Resposta/genética , Transdução de Sinais/efeitos dos fármacos , Tiocarbamatos/farmacologia , Tiocarbamatos/toxicidade , Tioureia/farmacologia , Tioureia/toxicidade
14.
Am J Pathol ; 167(3): 651-61, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16127146

RESUMO

The presence of mucus obstruction and neutrophil-predominant inflammation in several lung disorders, such as cystic fibrosis, suggests a relationship between neutrophils and excess mucus production. Mechanisms of human neutrophil elastase (HNE)-induced mucin secretion by well-differentiated normal human bronchial epithelial (NHBE) cells maintained in air/liquid interface culture were investigated. HNE increased mucin secretion in a concentration-dependent manner, with maximal stimulation (more than twofold) occurring within a short (15 minutes) time period. Mucins MUC 5 AC and MUC 5 B, but not MUC 2, were released in response to HNE. Stimulation of mucin secretion required partial elastase enzymatic activity and did not appear to involve a soluble product released by the cells. HNE-stimulated secretion involved activation of protein kinase C (PKC), as HNE exposure rapidly provoked PKC enzymatic activity that was attenuated by the general PKC inhibitors calphostin C and bisindoylmaleimide I. Of the different isoforms, PKCalpha, delta, zeta, lambda, iota, and epsilon were constitutively expressed in NHBE cells while PKCbeta, eta, and mu were PMA-inducible. PKCdelta was the only isoform to translocate from cytoplasm to membrane in response to HNE. Inhibition of PKCdelta attenuated HNE-mediated mucin secretion. The results suggest HNE stimulation of mucin release by human airway epithelial cells involves intracellular activation of PKC, specifically the delta isoform.


Assuntos
Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Elastase de Leucócito/farmacologia , Mucinas/metabolismo , Proteína Quinase C/fisiologia , Brônquios/citologia , Diferenciação Celular , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Proteína Quinase C-delta , Inibidores de Proteínas Quinases/farmacologia
15.
Am J Respir Crit Care Med ; 171(4): 305-14, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15531749

RESUMO

Chronic obstructive pulmonary disease is marked by alveolar enlargement and excess production of airway mucus. Acrolein, a component of cigarette smoke, increases mucin 5AC (MUC5AC), a prevalent airway mucin in NCI-H292 cells by transcriptional activation, but the signal transduction pathways involved in acrolein-induced MUC5AC expression are unknown. Acrolein depleted cellular glutathione at doses of 10 muM or greater, higher than those sufficient (0.03 muM) to increase MUC5AC mRNA, suggesting that MUC5AC expression was independent of oxidative stress. In contrast, acrolein increased MUC5AC mRNA levels by phosphorylating epidermal growth factor receptor (EGFR) and mitogen-activated protein kinase 3/2, or MAPK 3/2(ERK1/2). Pretreating the cells with an EGFR-neutralizing antibody, or a metalloproteinase inhibitor, decreased the acrolein-induced MUC5AC mRNA increase. Small, interfering RNA directed against ADAM17 or MMP9 inhibited the acrolein-induced MUC5AC mRNA increase. Acrolein increased the release and subsequent activation of pro-MMP9. Acrolein increased MMP9 and decreased tissue inhibitor of metalloproteinase 3 (TIMP3), an endogenous inhibitor of ADAM17, transcripts. Together, these data suggest that acrolein induces MUC5AC expression via an initial ligand-dependent activation of EGFR mediated by ADAM17 and MMP9. In addition, a prolonged effect of acrolein may be mediated by altering MMP9 and TIMP3 transcription.


Assuntos
Acroleína/farmacologia , Receptores ErbB/efeitos dos fármacos , Receptores ErbB/metabolismo , Metaloproteases/metabolismo , Mucinas/metabolismo , Western Blotting , Células Cultivadas , Glutationa/metabolismo , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mucina-5AC , Mucinas/efeitos dos fármacos , Mucinas/genética , Estresse Oxidativo/fisiologia , Reação em Cadeia da Polimerase/métodos , RNA Mensageiro/biossíntese , RNA Mensageiro/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
16.
Am J Physiol Gastrointest Liver Physiol ; 284(1): G46-56, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12388204

RESUMO

We have previously shown that PGE(2) and PGI(2) induce recovery of transepithelial resistance (TER) in ischemia-injured porcine ileal mucosa, associated with initial increases in Cl(-) secretion. We believe that the latter generates an osmotic gradient that stimulates resealing of tight junctions. Because of evidence implicating phosphatidylinositol 3-kinase (PI3K) in regulating tight junction assembly, we postulated that this signaling pathway is involved in PG-induced mucosal recovery. Porcine ileum was subjected to 45 min of ischemia, after which TER was monitored for a 180-min recovery period. Endogenous PG production was inhibited with indomethacin (5 microM). PGE(2) (1 microM) and PGI(2) (1 microM) stimulated recovery of TER, which was inhibited by serosal application of the osmotic agent urea (300 mosmol/kgH(2)O). The PI3K inhibitor wortmannin (10 nM) blocked recovery of TER in response to PGs or mucosal urea. Immunofluorescence imaging of recovering epithelium revealed that PGs restored occludin and zonula occludens-1 distribution to interepithelial junctions, and this pattern was disrupted by pretreatment with wortmannin. These experiments suggest that PGs stimulate recovery of paracellular resistance via a mechanism involving transepithelial osmotic gradients and PI3K-dependent restoration of tight junction protein distribution.


Assuntos
Epoprostenol/análogos & derivados , Íleo/metabolismo , Mucosa Intestinal/enzimologia , Isquemia/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/fisiologia , 16,16-Dimetilprostaglandina E2/farmacologia , Animais , Antiulcerosos/farmacologia , Impedância Elétrica , Epoprostenol/farmacologia , Feminino , Humanos , Mucosa Intestinal/ultraestrutura , Masculino , Proteínas de Membrana/metabolismo , Microscopia Eletrônica , Ocludina , Pressão Osmótica , Fosfoproteínas/metabolismo , Inibidores da Agregação Plaquetária/farmacologia , Recuperação de Função Fisiológica/fisiologia , Transdução de Sinais/efeitos dos fármacos , Suínos , Junções Íntimas/enzimologia , Ureia/farmacologia , Proteína da Zônula de Oclusão-1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA