Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 3(16)2018 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-30135315

RESUMO

Wilms' tumor 1 (WT1) is a critical transcriptional regulator of mesothelial cells during lung development but is downregulated in postnatal stages and adult lungs. We recently showed that WT1 is upregulated in both mesothelial cells and mesenchymal cells in the pathogenesis of idiopathic pulmonary fibrosis (IPF), a fatal fibrotic lung disease. Although WT1-positive cell accumulation leading to severe fibrotic lung disease has been studied, the role of WT1 in fibroblast activation and pulmonary fibrosis remains elusive. Here, we show that WT1 functions as a positive regulator of fibroblast activation, including fibroproliferation, myofibroblast transformation, and extracellular matrix (ECM) production. Chromatin immunoprecipitation experiments indicate that WT1 binds directly to the promoter DNA sequence of α-smooth muscle actin (αSMA) to induce myofibroblast transformation. In support, the genetic lineage tracing identifies WT1 as a key driver of mesothelial-to-myofibroblast and fibroblast-to-myofibroblast transformation. Importantly, the partial loss of WT1 was sufficient to attenuate myofibroblast accumulation and pulmonary fibrosis in vivo. Further, our coculture studies show that WT1 upregulation leads to non-cell autonomous effects on neighboring cells. Thus, our data uncovered a pathogenic role of WT1 in IPF by promoting fibroblast activation in the peripheral areas of the lung and as a target for therapeutic intervention.


Assuntos
Actinas/genética , Fibrose Pulmonar Idiopática/patologia , Miofibroblastos/patologia , Proteínas Repressoras/metabolismo , Proteínas WT1/metabolismo , Adulto , Animais , Bleomicina/toxicidade , Diferenciação Celular/genética , Células Cultivadas , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Fibrose , Regulação da Expressão Gênica , Técnicas de Introdução de Genes , Humanos , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/genética , Pulmão/citologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Masculino , Camundongos Transgênicos , Cultura Primária de Células , Regiões Promotoras Genéticas/genética
2.
J Physiol ; 596(8): 1397-1417, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29380370

RESUMO

KEY POINTS: The mechanisms by which bacteria alter endothelial cell phenotypes and programme inflammatory angiogenesis remain unclear. In lung endothelial cells, we demonstrate that toll-like receptor 4 (TLR4) signalling induces activation of forkhead box protein C2 (FOXC2), a transcriptional factor implicated in lymphangiogenesis and endothelial specification, in an extracellular signal-regulated kinase (ERK)-dependent manner. TLR4-ERK-FOXC2 signalling regulates expression of the Notch ligand DLL4 and signals inflammatory angiogenesis in vivo and in vitro. Our work reveals a novel link between endothelial immune signalling (TLR pathway) and a vascular transcription factor, FOXC2, that regulates embryonic vascular development. This mechanism is likely to be relevant to pathological angiogenesis complicating inflammatory diseases in humans. ABSTRACT: Endothelial cells (ECs) mediate a specific and robust immune response to bacteria in sepsis through the activation of toll-like receptor (TLR) signalling. The mechanisms by which bacterial ligands released during sepsis programme EC specification and altered angiogenesis remain unclear. We postulated that the forkhead box protein C2 (FOXC2) transcriptional factor directs EC cell-fate decisions and angiogenesis during TLR signalling. In human lung ECs, lipopolysaccharide (LPS) induced ERK phosphorylation, FOXC2, and delta-like 4 (DLL4, the master regulator of sprouting angiogenesis expression) in a TLR4-dependent manner. LPS-mediated ERK phosphorylation resulted in FOXC2-ERK protein ligation, ERK-dependent FOXC2 serine and threonine phosphorylation, and subsequent activation of DLL4 gene expression. Chemical inhibition of ERK or ERK-2 dominant negative transfection disrupted LPS-mediated FOXC2 phosphorylation and transcriptional activation of FOXC2. FOXC2-siRNA or ERK-inhibition attenuated LPS-induced DLL4 expression and angiogenic sprouting in vitro. In vivo, intraperitoneal LPS induced ERK and FOXC2 phosphorylation, FOXC2 binding to DLL4 promoter, and FOXC2/DLL4 expression in the lung. ERK-inhibition suppressed LPS-induced FOXC2 phosphorylation, FOXC2-DLL4 promoter binding, and induction of FOXC2 and DLL4 in mouse lung ECs. LPS induced aberrant retinal angiogenesis and DLL4 expression in neonatal mice, which was attenuated with ERK inhibition. FOXC2+/- mice treated with LPS showed a mitigated increase in FOXC2 and DLL4 compared to FOXC2+/+ mice. These data reveal a new mechanism (TLR4-ERK-FOXC2-DLL4) by which sepsis-induced EC TLR signalling programmes EC specification and altered angiogenesis.


Assuntos
Células Endoteliais/imunologia , Fatores de Transcrição Forkhead/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Neovascularização Fisiológica , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Ligação ao Cálcio , Diferenciação Celular , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lipopolissacarídeos/toxicidade , Pulmão/irrigação sanguínea , Pulmão/embriologia , Pulmão/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo
3.
J Clin Invest ; 125(5): 2021-31, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25866971

RESUMO

Epithelial cells that line the conducting airways provide the initial barrier and innate immune responses to the abundant particles, microbes, and allergens that are inhaled throughout life. The transcription factors SPDEF and FOXA3 are both selectively expressed in epithelial cells lining the conducting airways, where they regulate goblet cell differentiation and mucus production. Moreover, these transcription factors are upregulated in chronic lung disorders, including asthma. Here, we show that expression of SPDEF or FOXA3 in airway epithelial cells in neonatal mice caused goblet cell differentiation, spontaneous eosinophilic inflammation, and airway hyperresponsiveness to methacholine. SPDEF expression promoted DC recruitment and activation in association with induction of Il33, Csf2, thymic stromal lymphopoietin (Tslp), and Ccl20 transcripts. Increased Il4, Il13, Ccl17, and Il25 expression was accompanied by recruitment of Th2 lymphocytes, group 2 innate lymphoid cells, and eosinophils to the lung. SPDEF was required for goblet cell differentiation and pulmonary Th2 inflammation in response to house dust mite (HDM) extract, as both were decreased in neonatal and adult Spdef(-/-) mice compared with control animals. Together, our results indicate that SPDEF causes goblet cell differentiation and Th2 inflammation during postnatal development and is required for goblet cell metaplasia and normal Th2 inflammatory responses to HDM aeroallergen.


Assuntos
Antígenos de Dermatophagoides/toxicidade , Células Epiteliais/metabolismo , Células Caliciformes/fisiologia , Pulmão/imunologia , Proteínas Proto-Oncogênicas c-ets/fisiologia , Eosinofilia Pulmonar/imunologia , Células Th2/imunologia , Fatores Etários , Animais , Animais Recém-Nascidos , Diferenciação Celular , Quimiocina CCL20/biossíntese , Quimiocina CCL20/genética , Quimiotaxia de Leucócito , Citocinas/antagonistas & inibidores , Citocinas/biossíntese , Citocinas/genética , Células Dendríticas/imunologia , Eosinófilos/fisiologia , Fator 3-gama Nuclear de Hepatócito/fisiologia , Interleucinas/biossíntese , Interleucinas/genética , Metaplasia , Cloreto de Metacolina/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-ets/genética , Eosinofilia Pulmonar/etiologia , Eosinofilia Pulmonar/metabolismo , Eosinofilia Pulmonar/patologia , Proteínas Recombinantes de Fusão/metabolismo , Transgenes , Linfopoietina do Estroma do Timo
4.
Am J Physiol Lung Cell Mol Physiol ; 306(8): L726-35, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24508732

RESUMO

A number of growth factors and signaling pathways regulate matrix deposition and fibroblast proliferation in the lung. The epidermal growth factor receptor (EGFR) family of receptors and the transforming growth factor-ß (TGF-ß) family are active in diverse biological processes and are central mediators in the initiation and maintenance of fibrosis in many diseases. Transforming growth factor-α (TGF-α) is a ligand for the EGFR, and doxycycline (Dox)-inducible transgenic mice conditionally expressing TGF-α specifically in the lung epithelium develop progressive fibrosis accompanied with cachexia, changes in lung mechanics, and marked pleural thickening. Although recent studies demonstrate that EGFR activation modulates the fibroproliferative effects involved in the pathogenesis of TGF-ß induced pulmonary fibrosis, in converse, the direct role of EGFR induction of the TGF-ß pathway in the lung is unknown. The αvß6 integrin is an important in vivo activator of TGF-ß activation in the lung. Immunohistochemical analysis of αvß6 protein expression and bronchoalveolar analysis of TGF-ß pathway signaling indicates activation of the αvß6/TGF-ß pathway only at later time points after lung fibrosis was already established in the TGF-α model. To determine the contribution of the αvß6/TGF-ß pathway on the progression of established fibrotic disease, TGF-α transgenic mice were administered Dox for 4 wk, which leads to extensive fibrosis; these mice were then treated with a function-blocking anti-αvß6 antibody with continued administration of Dox for an additional 4 wk. Compared with TGF-α transgenic mice treated with control antibody, αvß6 inhibition significantly attenuated pleural thickening and altered the decline in lung mechanics. To test the effects of genetic loss of the ß6 integrin, TGF-α transgenic mice were mated with ß6-null mice and the degree of fibrosis was compared in adult mice following 8 wk of Dox administration. Genetic ablation of the ß6 integrin attenuated histological and physiological changes in the lungs of TGF-α transgenic mice although a significant degree of fibrosis still developed. In summary, inhibition of the ß6 integrin led to a modest, albeit significant, effect on pleural thickening and lung function decline observed with TGF-α-induced pulmonary fibrosis. These data support activation of the αvß6/TGF-ß pathway as a secondary effect contributing to TGF-α-induced pleural fibrosis and suggest a complex contribution of multiple mediators to the maintenance of progressive fibrosis in the lung.


Assuntos
Integrinas/antagonistas & inibidores , Fibrose Pulmonar/patologia , Fator de Crescimento Transformador alfa/farmacologia , Animais , Antibacterianos/toxicidade , Anticorpos Neutralizantes , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Lavagem Broncoalveolar , Colágeno , Doxiciclina/toxicidade , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Técnicas Imunoenzimáticas , Integrinas/genética , Integrinas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta/farmacologia , Uteroglobina/fisiologia
5.
Am J Respir Crit Care Med ; 189(3): 301-13, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24392884

RESUMO

RATIONALE: Goblet cell metaplasia accompanies common pulmonary disorders that are prone to recurrent viral infections. Mechanisms regulating both goblet cell metaplasia and susceptibility to viral infection associated with chronic lung diseases are incompletely understood. OBJECTIVES: We sought to identify the role of the transcription factor FOXA3 in regulation of goblet cell metaplasia and pulmonary innate immunity. METHODS: FOXA3 was identified in airways from patients with asthma and chronic obstructive pulmonary disease. We produced transgenic mice conditionally expressing Foxa3 in airway epithelial cells and developed human bronchial epithelial cells expressing Foxa3. Foxa3-regulated genes were identified by immunostaining, Western blotting, and RNA analysis. Direct binding of FOXA3 to target genes was identified by chromatin immunoprecipitation sequencing correlated with RNA sequencing. MEASUREMENTS AND MAIN RESULTS: FOXA3 was highly expressed in airway goblet cells from patients with asthma and chronic obstructive pulmonary disease. FOXA3 was induced by either IL-13 or rhinovirus. Foxa3 induced goblet cell metaplasia and enhanced expression of a network of genes mediating mucus production. Paradoxically, FOXA3 inhibited rhinovirus-induced IFN production, IRF-3 phosphorylation, and IKKε expression and inhibited viral clearance and expression of genes required for antiviral defenses, including MDA5, RIG-I, TLR3, IRF7/9, and nuclear factor-κB. CONCLUSIONS: FOXA3 induces goblet cell metaplasia in response to infection or Th2 stimulation. Suppression of IFN signaling by FOXA3 provides a plausible mechanism that may serve to limit ongoing Th1 inflammation during the resolution of acute viral infection; however, inhibition of innate immunity by FOXA3 may contribute to susceptibility to viral infections associated with chronic lung disorders accompanied by chronic goblet cell metaplasia.


Assuntos
Asma/metabolismo , Células Caliciformes/patologia , Fator 3-gama Nuclear de Hepatócito/metabolismo , Imunidade Inata/fisiologia , Infecções por Picornaviridae/imunologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Animais , Asma/complicações , Asma/imunologia , Asma/patologia , Biomarcadores/metabolismo , Western Blotting , Imunoprecipitação da Cromatina , Suscetibilidade a Doenças , Células Caliciformes/imunologia , Células Caliciformes/metabolismo , Fator 3-gama Nuclear de Hepatócito/imunologia , Humanos , Interferons/metabolismo , Metaplasia , Camundongos , Camundongos Transgênicos , Infecções por Picornaviridae/etiologia , Doença Pulmonar Obstrutiva Crônica/complicações , Doença Pulmonar Obstrutiva Crônica/imunologia , Doença Pulmonar Obstrutiva Crônica/patologia , Rhinovirus , Análise de Sequência de RNA , Equilíbrio Th1-Th2
6.
PLoS One ; 9(1): e86536, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24475138

RESUMO

Pulmonary fibrosis is often triggered by an epithelial injury resulting in the formation of fibrotic lesions in the lung, which progress to impair gas exchange and ultimately cause death. Recent clinical trials using drugs that target either inflammation or a specific molecule have failed, suggesting that multiple pathways and cellular processes need to be attenuated for effective reversal of established and progressive fibrosis. Although activation of MAPK and PI3K pathways have been detected in human fibrotic lung samples, the therapeutic benefits of in vivo modulation of the MAPK and PI3K pathways in combination are unknown. Overexpression of TGFα in the lung epithelium of transgenic mice results in the formation of fibrotic lesions similar to those found in human pulmonary fibrosis, and previous work from our group shows that inhibitors of either the MAPK or PI3K pathway can alter the progression of fibrosis. In this study, we sought to determine whether simultaneous inhibition of the MAPK and PI3K signaling pathways is a more effective therapeutic strategy for established and progressive pulmonary fibrosis. Our results showed that inhibiting both pathways had additive effects compared to inhibiting either pathway alone in reducing fibrotic burden, including reducing lung weight, pleural thickness, and total collagen in the lungs of TGFα mice. This study demonstrates that inhibiting MEK and PI3K in combination abolishes proliferative changes associated with fibrosis and myfibroblast accumulation and thus may serve as a therapeutic option in the treatment of human fibrotic lung disease where these pathways play a role.


Assuntos
Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Inibidores de Fosfoinositídeo-3 Quinase , Fibrose Pulmonar/tratamento farmacológico , Análise de Variância , Animais , Benzimidazóis/farmacologia , Western Blotting , Quimioterapia Combinada , Gonanos/farmacologia , Imuno-Histoquímica , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de RNA , Fator de Crescimento Transformador alfa/metabolismo
7.
Am J Respir Cell Mol Biol ; 49(5): 845-54, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23795648

RESUMO

Pulmonary surfactant protein-C (SP-C) gene-targeted mice (Sftpc(-/-)) develop progressive lung inflammation and remodeling. We hypothesized that SP-C deficiency reduces the ability to suppress repetitive inflammatory injury. Sftpc(+/+) and Sftpc(-/-) mice given three doses of bacterial LPS developed airway and airspace inflammation, which was more intense in the Sftpc(-/-) mice at 3 and 5 days after the final dose. Compared with Sftpc(+/+)mice, inflammatory injury persisted in the lungs of Sftpc(-/-) mice 30 days after the final LPS challenge. Sftpc(-/-) mice showed LPS-induced airway goblet cell hyperplasia with increased detection of Sam pointed Ets domain and FoxA3 transcription factors. Sftpc(-/-) type II alveolar epithelial cells had increased cytokine expression after LPS exposure relative to Sftpc(+/+) cells, indicating that type II cell dysfunction contributes to inflammatory sensitivity. Microarray analyses of isolated type II cells identified a pattern of enhanced expression of inflammatory genes consistent with an intrinsic low-level inflammation resulting from SP-C deficiency. SP-C-containing clinical surfactant extract (Survanta) or SP-C/phospholipid vesicles blocked LPS signaling through the LPS receptor (Toll-like receptor [TLR] 4/CD14/MD2) in human embryonic kidney 293T cells, indicating that SP-C blocks LPS-induced cytokine production by a TLR4-dependent mechanism. Phospholipid vesicles alone did not modify the TLR4 response. In vivo deficiency of SP-C leads to inflammation, increased cytokine production by type II cells, and persistent inflammation after repetitive LPS stimulation.


Assuntos
Endotoxinas , Pulmão/metabolismo , Peptídeos/deficiência , Pneumonia/metabolismo , Células Epiteliais Alveolares/imunologia , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/patologia , Animais , Produtos Biológicos/farmacologia , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Células Caliciformes/imunologia , Células Caliciformes/metabolismo , Células Caliciformes/patologia , Células HEK293 , Fator 3-gama Nuclear de Hepatócito/metabolismo , Humanos , Hiperplasia , Imunidade Inata , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Receptores de Lipopolissacarídeos/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Peptídeos/genética , Pneumonia/induzido quimicamente , Pneumonia/genética , Pneumonia/imunologia , Pneumonia/patologia , Proteínas Proto-Oncogênicas c-ets/metabolismo , Proteína C Associada a Surfactante Pulmonar , Transdução de Sinais , Fatores de Tempo , Receptor 4 Toll-Like/metabolismo
8.
Respir Res ; 14: 19, 2013 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-23399055

RESUMO

BACKGROUND: Individuals with deficiencies of pulmonary surfactant protein C (SP-C) develop interstitial lung disease (ILD) that is exacerbated by viral infections including respiratory syncytial virus (RSV). SP-C gene targeted mice (Sftpc -/-) lack SP-C, develop an ILD-like disease and are susceptible to infection with RSV. METHODS: In order to determine requirements for correction of RSV induced injury we have generated compound transgenic mice where SP-C expression can be induced on the Sftpc -/- background (SP-C/Sftpc -/-) by the administration of doxycycline (dox). The pattern of induced SP-C expression was determined by immunohistochemistry and processing by Western blot analysis. Tissue and cellular inflammation was measured following RSV infection and the RSV-induced cytokine response of isolated Sftpc +/+ and -/- type II cells determined. RESULTS: After 5 days of dox administration transgene SP-C mRNA expression was detected by RT-PCR in the lungs of two independent lines of bitransgenic SP-C/Sftpc -/- mice (lines 55.3 and 54.2). ProSP-C was expressed in the lung, and mature SP-C was detected by Western blot analysis of the lavage fluid from both lines of SP-C/Sftpc -/- mice. Induced SP-C expression was localized to alveolar type II cells by immunostaining with an antibody to proSP-C. Line 55.3 SP-C/Sftpc -/- mice were maintained on or off dox for 7 days and infected with 2.6x107 RSV pfu. On day 3 post RSV infection total inflammatory cell counts were reduced in the lavage of dox treated 55.3 SP-C/Sftpc -/- mice (p = 0.004). The percentage of neutrophils was reduced (p = 0.05). The viral titers of lung homogenates from dox treated 55.3 SP-C/Sftpc -/- mice were decreased relative to 55.3 SP-C/Sftpc -/- mice without dox (p = 0.01). The cytokine response of Sftpc -/- type II cells to RSV was increased over that of Sftpc +/+ cells. CONCLUSIONS: Transgenic restoration of SP-C reduced inflammation and improved viral clearance in the lungs of SP-C deficient mice. The loss of SP-C in alveolar type II cells compromises their response to infection. These findings show that the restoration of SP-C in Sftpc -/- mice in response to RSV infection is a useful model to determine parameters for therapeutic intervention.


Assuntos
Lesão Pulmonar/metabolismo , Proteína C Associada a Surfactante Pulmonar/genética , Infecções por Vírus Respiratório Sincicial/genética , Vírus Sinciciais Respiratórios , Animais , Células Cultivadas , Regulação para Baixo/genética , Lesão Pulmonar/genética , Lesão Pulmonar/prevenção & controle , Camundongos , Camundongos da Linhagem 129 , Camundongos Transgênicos , Proteína C Associada a Surfactante Pulmonar/biossíntese , Infecções por Vírus Respiratório Sincicial/metabolismo , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Carga Viral/métodos
9.
Proc Natl Acad Sci U S A ; 109(41): 16630-5, 2012 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-23012424

RESUMO

Airway mucus plays a critical role in clearing inhaled toxins, particles, and pathogens. Diverse toxic, inflammatory, and infectious insults induce airway mucus secretion and goblet cell metaplasia to preserve airway sterility and homeostasis. However, goblet cell metaplasia, mucus hypersecretion, and airway obstruction are integral features of inflammatory lung diseases, including asthma, chronic obstructive lung disease, and cystic fibrosis, which cause an immense burden of morbidity and mortality. These chronic lung diseases are united by susceptibility to microbial colonization and recurrent airway infections. Whether these twinned phenomena (mucous metaplasia, compromised host defenses) are causally related has been unclear. Here, we demonstrate that SAM pointed domain ETS factor (SPDEF) was induced by rhinoviral infection of primary human airway cells and that cytoplasmic activities of SPDEF, a transcriptional regulator of airway goblet cell metaplasia, inhibited Toll-like receptor (TLR) activation of epithelial cells. SPDEF bound to and inhibited activities of TLR signaling adapters, MyD88 and TRIF, inhibiting MyD88-induced cytokine production and TRIF-induced interferon ß production. Conditional expression of SPDEF in airway epithelial cells in vivo inhibited LPS-induced neutrophilic infiltration and bacterial clearance. SPDEF-mediated inhibition of both TLR and type I interferon signaling likely protects the lung against inflammatory damage when inciting stimuli are not eradicated. Present findings provide, at least in part, a molecular explanation for increased susceptibility to infection in lung diseases associated with mucous metaplasia and a mechanism by which patients with florid mucous metaplasia may tolerate microbial burdens that are usually associated with fulminant inflammatory disease in normal hosts.


Assuntos
Células Epiteliais/metabolismo , Proteínas Proto-Oncogênicas c-ets/metabolismo , Mucosa Respiratória/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Antibacterianos/farmacologia , Western Blotting , Doxiciclina/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Expressão Gênica/efeitos dos fármacos , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Interleucina-13/farmacologia , Lipopolissacarídeos/farmacologia , Pneumopatias/tratamento farmacológico , Pneumopatias/metabolismo , Pneumopatias/patologia , Metaplasia , Camundongos , Microscopia Confocal , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas c-ets/genética , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rhinovirus/fisiologia , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
10.
Respir Res ; 13: 51, 2012 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-22726462

RESUMO

BACKGROUND: Resistin-like molecule alpha or found in inflammatory zone protein (Fizz1) is increased in pulmonary epithelial cells and also in limited amounts by other lung cells during various lung injuries and fibrosis. However, the direct role of Fizz1 produced in the pulmonary epithelium has not been determined. METHODS: Fizz1 Transgenic mice (CCSP/Fizz1) were generated that overexpress Fizz1 in the lung epithelium under the control of a doxycycline (Dox) inducible lung epithelial cell specific promoter Scgb1a1 (Clara cell secretory protein, CCSP). Histology and FACS analysis of lung cells were used to identify the direct effects of Fizz1 in the transgenic mice (Dox treated) when compared with control (CCSP/-) mice. Intratracheal bleomycin sulfate or silica in saline and saline alone were used to study the role of Fizz1 during bleomycin- and silica-induced pulmonary fibrosis in CCSP/Fizz1 and CCSP/- mice. Weight change, pulmonary inflammation, and fibrosis were assessed 10 days post bleomycin or 28 days post silica challenge. RESULTS: When CCSP/Fizz1 mice were fed Dox food, elevated Fizz1 protein was detected in lung homogenates by western blot. Lungs of mice in which Fizz1 was induced in the epithelium contained increased lung cells staining for CD11c and F4/80 by FACS analysis consistent with increased dendritic cells however, no changes were observed in the percentage of interstitial macrophages compared to CCSP/- controls. No significant changes were found in the lung histology of CCSP/Fizz1 mice after up to 8 weeks of overexpression compared to CCSP/- controls. Overexpression of Fizz1 prior to challenge or following challenge with bleomycin or silica did not significantly alter airway inflammation or fibrosis compared to control mice. CONCLUSIONS: The current study demonstrates that epithelial cell derived Fizz1 is sufficient to increase the bone-marrow derived dendritic cells in the lungs, but it is not sufficient to cause lung fibrosis or alter chemical or particle-induced fibrosis.


Assuntos
Movimento Celular/fisiologia , Células Dendríticas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Pulmão/metabolismo , Pulmão/patologia , Fibrose Pulmonar , Animais , Células Dendríticas/patologia , Feminino , Camundongos , Camundongos Transgênicos , Fibrose Pulmonar/etiologia , Fibrose Pulmonar/patologia
11.
J Appl Physiol (1985) ; 111(6): 1760-7, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21903885

RESUMO

Airway hyperreactivity (AHR) and remodeling are cardinal features of asthma and chronic obstructive pulmonary disease. New therapeutic targets are needed as some patients are refractory to current therapies and develop progressive airway remodeling and worsening AHR. The mammalian target of rapamycin (mTOR) is a key regulator of cellular proliferation and survival. Treatment with the mTOR inhibitor rapamycin inhibits inflammation and AHR in allergic asthma models, but it is unclear if rapamycin can directly inhibit airway remodeling and AHR, or whether its therapeutic effects are entirely mediated through immunosuppression. To address this question, we utilized transforming growth factor-α (TGF-α) transgenic mice null for the transcription factor early growth response-1 (Egr-1) (TGF-α Tg/Egr-1(ko/ko) mice). These mice develop airway smooth muscle thickening and AHR in the absence of altered lung inflammation, as previously reported. In this study, TGF-α Tg/Egr-1(ko/ko) mice lost body weight and developed severe AHR after 3 wk of lung-specific TGF-α induction. Rapamycin treatment prevented body weight loss, airway wall thickening, abnormal lung mechanics, and increases in airway resistance to methacholine after 3 wk of TGF-α induction. Increases in tissue damping and airway elastance were also attenuated in transgenic mice treated with rapamycin. TGF-α/Egr-1(ko/ko) mice on doxycycline for 8 wk developed severe airway remodeling. Immunostaining for α-smooth muscle actin and morphometric analysis showed that rapamycin treatment prevented airway smooth muscle thickening around small airways. Pentachrome staining, assessments of lung collagen and fibronectin mRNA levels, indicated that rapamycin also attenuated fibrotic pathways induced by TGF-α expression for 8 wk. Thus rapamycin reduced airway remodeling and AHR, demonstrating an important role for mTOR signaling in TGF-α-induced/EGF receptor-mediated reactive airway disease.


Assuntos
Remodelação das Vias Aéreas/efeitos dos fármacos , Hiper-Reatividade Brônquica/tratamento farmacológico , Pneumopatias/tratamento farmacológico , Pneumopatias/fisiopatologia , Sirolimo/farmacologia , Remodelação das Vias Aéreas/genética , Remodelação das Vias Aéreas/fisiologia , Animais , Hiper-Reatividade Brônquica/genética , Hiper-Reatividade Brônquica/fisiopatologia , Proteína 1 de Resposta de Crescimento Precoce/deficiência , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/fisiologia , Receptores ErbB/fisiologia , Pneumopatias/genética , Pneumopatias/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/fisiologia , Fator de Crescimento Transformador alfa/genética , Fator de Crescimento Transformador alfa/fisiologia
12.
Am J Physiol Lung Cell Mol Physiol ; 300(3): L414-21, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21224214

RESUMO

Increases in the epidermal growth factor receptor (EGFR) have been associated with the severity of airway thickening in chronic asthmatic subjects, and EGFR signaling is induced by asthma-related cytokines and inflammation. The goal of this study was to determine the role of EGFR signaling in a chronic allergic model of asthma and specifically in epithelial cells, which are increasingly recognized as playing an important role in asthma. EGFR activation was assessed in mice treated with intranasal house dust mite (HDM) for 3 wk. EGFR signaling was inhibited in mice treated with HDM for 6 wk, by using either the drug erlotinib or a genetic approach that utilizes transgenic mice expressing a mutant dominant negative epidermal growth factor receptor in the lung epithelium (EGFR-M mice). Airway hyperreactivity (AHR) was assessed by use of a flexiVent system after increasing doses of nebulized methacholine. Airway smooth muscle (ASM) thickening was measured by morphometric analysis. Sensitization to HDM (IgG and IgE), inflammatory cells, and goblet cell changes were also assessed. Increased EGFR activation was detected in HDM-treated mice, including in bronchiolar epithelial cells. In mice exposed to HDM for 6 wk, AHR and ASM thickening were reduced after erlotinib treatment and in EGFR-M mice. Sensitization to HDM and inflammatory cell counts were similar in all groups, except neutrophil counts, which were lower in the EGFR-M mice. Goblet cell metaplasia with HDM treatment was reduced by erlotinib, but not in EGFR-M transgenic mice. This study demonstrates that EGFR signaling, especially in the airway epithelium, plays an important role in mediating AHR and remodeling in a chronic allergic asthma model.


Assuntos
Remodelação das Vias Aéreas/fisiologia , Asma/fisiopatologia , Hiper-Reatividade Brônquica/complicações , Células Epiteliais/enzimologia , Receptores ErbB/metabolismo , Transdução de Sinais , Animais , Asma/complicações , Asma/parasitologia , Asma/patologia , Hiper-Reatividade Brônquica/parasitologia , Hiper-Reatividade Brônquica/patologia , Hiper-Reatividade Brônquica/fisiopatologia , Doença Crônica , Modelos Animais de Doenças , Ativação Enzimática , Células Epiteliais/patologia , Receptores ErbB/antagonistas & inibidores , Células Caliciformes/patologia , Inflamação/complicações , Inflamação/patologia , Pulmão/parasitologia , Pulmão/patologia , Pulmão/fisiopatologia , Metaplasia , Camundongos , Músculo Liso/patologia , Pyroglyphidae/fisiologia
13.
Am J Respir Cell Mol Biol ; 44(2): 175-84, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20348208

RESUMO

Surfactant protein A (SP-A) mediates innate immune cell responses to LPS, a cell wall component of gram-negative bacteria that is found ubiquitously in the environment and is associated with adverse health effects. Inhaled LPS induces lung inflammation and increases airway responsiveness (AR). However, the role of SP-A in mediating LPS-induced AR is not well-defined. Nitric oxide (NO) is described as a potent bronchodilator, and previous studies showed that SP-A modulates the LPS-induced production of NO. Hence, we tested the hypothesis that increased AR, observed in response to aerosolized LPS exposure, would be significantly reduced in an SP-A-deficient condition. Wild-type (WT) and SP-A null (SP-A(-/-)) mice were challenged with aerosolized LPS. Results indicate that despite similar inflammatory indices, LPS-treated SP-A(-/-) mice had attenuated AR after methacholine challenge, compared with WT mice. The attenuated AR could not be attributed to inherent differences in SP-D concentrations or airway smooth muscle contractile and relaxation properties, because these measures were similar between WT and SP-A(-/-) mice. LPS-treated SP-A(-/-) mice, however, had elevated nitrite concentrations, inducible nitric oxide synthase (iNOS) expression, and NOS activity in their lungs. Moreover, the administration of the iNOS-specific inhibitor 1400W completely abrogated the attenuated AR. Thus, when exposed to aerosolized LPS, SP-A(-/-) mice demonstrate a relative airway hyporesponsiveness that appears to be mediated at least partly via an iNOS-dependent mechanism. These findings may have clinical significance, because recent studies reported associations between surfactant protein polymorphisms and a variety of lung diseases.


Assuntos
Lipopolissacarídeos/farmacologia , Pulmão/imunologia , Pulmão/fisiopatologia , Óxido Nítrico/fisiologia , Proteína A Associada a Surfactante Pulmonar/deficiência , Animais , Imunidade Inata , Pulmão/efeitos dos fármacos , Cloreto de Metacolina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/metabolismo , Proteína A Associada a Surfactante Pulmonar/genética , Proteína A Associada a Surfactante Pulmonar/imunologia , Proteína A Associada a Surfactante Pulmonar/fisiologia , Proteína D Associada a Surfactante Pulmonar/metabolismo
14.
Cell Cycle ; 9(14): 2769-76, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20676040

RESUMO

Pulmonary fibrosis complicates a number of disease processes and leads to substantial morbidity and mortality. Idiopathic pulmonary fibrosis (IPF) is perhaps the most pernicious and enigmatic form of the greater problem of lung fibrogenesis with a median survival of three years from diagnosis in affected patients. In this review, we will focus on the pathology of IPF as a model of pulmonary fibrotic processes, review possible cellular mechanisms, review current treatment approaches and review two transgenic mouse models of lung fibrosis to provide insight into processes that cause lung fibrosis. We will also summarize the potential utility of signaling pathway inhibitors as a future treatment in pulmonary fibrosis. Finally, we will present data demonstrating a minimal contribution of epithelial-mesenchymal transition in the development of fibrotic lesions in the transforming growth factor-alpha transgenic model of lung fibrosis.


Assuntos
Células Epiteliais/metabolismo , Fibrose Pulmonar/etiologia , Transdução de Sinais , Animais , Modelos Animais de Doenças , Células Epiteliais/citologia , Receptores ErbB/metabolismo , Humanos , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/terapia , Proteína C Associada a Surfactante Pulmonar/genética , Proteína C Associada a Surfactante Pulmonar/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
15.
Am J Pathol ; 176(2): 679-86, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20042669

RESUMO

Transforming growth factor-alpha (TGFalpha) is a ligand for the epidermal growth factor receptor (EGFR). EGFR activation is associated with fibroproliferative processes in human lung disease and animal models of pulmonary fibrosis. EGFR signaling activates several intracellular signaling pathways including phosphatidylinositol 3'-kinase (PI3K). We previously showed that induction of lung-specific TGFalpha expression in transgenic mice caused progressive pulmonary fibrosis over a 4-week period. The increase in levels of phosphorylated Akt, detected after 1 day of doxycycline-induced TGFalpha expression, was blocked by treatment with the PI3K inhibitor, PX-866. Daily administration of PX-866 during TGFalpha induction prevented increases in lung collagen and airway resistance as well as decreases in lung compliance. Treatment of mice with oral PX-866 4 weeks after the induction of TGFalpha prevented additional weight loss and further increases in total collagen, and attenuated changes in pulmonary mechanics. These data show that PI3K is activated in TGFalpha/EGFR-mediated pulmonary fibrosis and support further studies to determine the role of PI3K activation in human lung fibrotic disease, which could be amenable to targeted therapy.


Assuntos
Gonanos/farmacologia , Gonanos/uso terapêutico , Inibidores de Fosfoinositídeo-3 Quinase , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/prevenção & controle , Fator de Crescimento Transformador alfa , Administração Oral , Animais , Progressão da Doença , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Gonanos/administração & dosagem , Camundongos , Camundongos Transgênicos , Proteína Oncogênica v-akt/metabolismo , Fosforilação/efeitos dos fármacos , Uteroglobina/genética
16.
Expert Opin Ther Targets ; 14(2): 117-30, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20055712

RESUMO

IMPORTANCE OF THE FIELD: CF airway mucus can be infected by opportunistic microorganisms, notably Pseudomonas aeruginosa. Once organisms are established as biofilms, even the most potent antibiotics have little effect on their viability, especially during late-stage chronic infections. Better understanding of the mechanisms used by P. aeruginosa to circumvent host defenses and therapeutic intervention strategies is critical for advancing novel treatment strategies. AREAS COVERED IN THIS REVIEW: Inflammatory injury in CF lung, role of neutrophils in pathogenesis, P. aeruginosa biofilms, mucoidy and its relationship with poor airway oxygenation, mechanisms by which P. aeruginosa biofilms in the CF airway can be killed. WHAT THE READER WILL GAIN: An understanding of the processes that P. aeruginosa undergoes during CF airway disease and clues to better treat such infections in future. TAKE HOME MESSAGE: The course of CF airway disease is a process involving host and microbial factors that often dictate frequency of pulmonary exacerbations, thus affecting the overall course. In the past decade significant discoveries have been made regarding the pathogenic processes used by P. aeruginosa to bypass the immune system. Many new and exciting features of P. aeruginosa now illuminate weaknesses in the organism that may render it susceptible to inexpensive compounds that force its own destruction.


Assuntos
Biofilmes , Fibrose Cística/microbiologia , Infecções por Pseudomonas/etiologia , Pseudomonas aeruginosa/fisiologia , Fibrose Cística/patologia , Fímbrias Bacterianas/fisiologia , Flagelos/fisiologia , Humanos , Pulmão/microbiologia , Pulmão/patologia , Neutrófilos/fisiologia , Infecções por Pseudomonas/tratamento farmacológico , Percepção de Quorum , Nitrito de Sódio/metabolismo
17.
J Clin Invest ; 119(10): 2914-24, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19759516

RESUMO

Various acute and chronic inflammatory stimuli increase the number and activity of pulmonary mucus-producing goblet cells, and goblet cell hyperplasia and excess mucus production are central to the pathogenesis of chronic pulmonary diseases. However, little is known about the transcriptional programs that regulate goblet cell differentiation. Here, we show that SAM-pointed domain-containing Ets-like factor (SPDEF) controls a transcriptional program critical for pulmonary goblet cell differentiation in mice. Initial cell-lineage-tracing analysis identified nonciliated secretory epithelial cells, known as Clara cells, as the progenitors of goblet cells induced by pulmonary allergen exposure in vivo. Furthermore, in vivo expression of SPDEF in Clara cells caused rapid and reversible goblet cell differentiation in the absence of cell proliferation. This was associated with enhanced expression of genes regulating goblet cell differentiation and protein glycosylation, including forkhead box A3 (Foxa3), anterior gradient 2 (Agr2), and glucosaminyl (N-acetyl) transferase 3, mucin type (Gcnt3). Consistent with these findings, levels of SPDEF and FOXA3 were increased in mouse goblet cells after sensitization with pulmonary allergen, and the proteins were colocalized in goblet cells lining the airways of patients with chronic lung diseases. Deletion of the mouse Spdef gene resulted in the absence of goblet cells in tracheal/laryngeal submucosal glands and in the conducting airway epithelium after pulmonary allergen exposure in vivo. These data show that SPDEF plays a critical role in regulating a transcriptional network mediating the goblet cell differentiation and mucus hyperproduction associated with chronic pulmonary disorders.


Assuntos
Diferenciação Celular/fisiologia , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Células Caliciformes/fisiologia , Muco/metabolismo , Proteínas Proto-Oncogênicas c-ets/metabolismo , Mucosa Respiratória/citologia , Animais , Linhagem Celular , Perfilação da Expressão Gênica , Células Caliciformes/citologia , Fator 3-gama Nuclear de Hepatócito/genética , Fator 3-gama Nuclear de Hepatócito/metabolismo , Humanos , Pulmão/citologia , Pulmão/fisiologia , Pneumopatias/metabolismo , Pneumopatias/patologia , Camundongos , Camundongos Knockout , Mucinas/biossíntese , Mucinas/genética , Mucoproteínas/genética , Mucoproteínas/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Oncogênicas , Proteínas Proto-Oncogênicas c-ets/genética , Mucosa Respiratória/fisiologia
18.
Am J Respir Crit Care Med ; 180(9): 834-45, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19661247

RESUMO

RATIONALE: Induced mainly by cigarette smoking, chronic obstructive pulmonary disease (COPD) is a global public health problem characterized by progressive difficulty in breathing and increased mucin production. Previously, we reported that acrolein levels found in COPD sputum could activate matrix metalloproteinase-9 (MMP9). OBJECTIVES: To determine whether acrolein increases expression and activity of MMP14, a critical membrane-bound endopeptidase that can initial a MMP-activation cascade. METHODS: MMP14 activity and adduct formation were measured following direct acrolein treatment. MMP14 expression and activity was measured in human airway epithelial cells. MMP14 immunohistochemistry was performed with COPD tissue, and in acrolein- or tobacco-exposed mice. MEASUREMENTS AND MAIN RESULTS: In a cell-free system, acrolein, in concentrations equal to those found in COPD sputum, directly adducted cysteine 319 in the MMP14 hemopexin-like domain and activated MMP14. In cells, acrolein increased MMP14 activity, which was inhibited by a proprotein convertase inhibitor, hexa-d-arginine. In the airway epithelium of COPD subjects, immunoreactive MMP14 protein increased. In mouse lung, acrolein or tobacco smoke increased lung MMP14 activity and protein. In cells, acrolein-induced MMP14 transcripts were inhibited by an epidermal growth factor receptor (EGFR) neutralizing antibody, EGFR kinase inhibitor, metalloproteinase inhibitor, or mitogen-activated protein kinase (MAPK) 3/2 or MAPK8 inhibitors, but not a MAPK14 inhibitor. Decreasing the MMP14 protein and activity in vitro by small interfering (si)RNA to MMP14 diminished the acrolein-induced MUC5AC transcripts. In acrolein-exposed mice or transgenic mice with lung-specific transforming growth factor-alpha (an EGFR ligand) expression, lung MMP14 and MUC5AC levels increased and these effects were inhibited by a EGFR inhibitor, erlotinib. CONCLUSIONS: Taken together, these findings implicate acrolein-induced MMP14 expression and activity in mucin production in COPD.


Assuntos
Metaloproteinase 14 da Matriz/metabolismo , Mucinas/biossíntese , Mucosa Respiratória/metabolismo , Acroleína/metabolismo , Animais , Ativação Enzimática , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Cloridrato de Erlotinib , Regulação Enzimológica da Expressão Gênica , Humanos , Pulmão/enzimologia , Pulmão/metabolismo , Camundongos , Mucinas/metabolismo , Inibidores de Proteínas Quinases/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Quinazolinas/metabolismo , Mucosa Respiratória/ultraestrutura
19.
Am J Physiol Lung Cell Mol Physiol ; 297(1): L64-72, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19304906

RESUMO

Patients with mutations in the pulmonary surfactant protein C (SP-C) gene develop interstitial lung disease and pulmonary exacerbations associated with viral infections including respiratory syncytial virus (RSV). Pulmonary infection with RSV caused more severe interstitial thickening, air space consolidation, and goblet cell hyperplasia in SP-C-deficient (Sftpc(-/-)) mice compared with SP-C replete mice. The RSV-induced pathology resolved more slowly in Sftpc(-/-) mice with lung inflammation persistent up to 30 days postinfection. Polymorphonuclear leukocyte and macrophage counts were increased in the bronchoalveolar lavage (BAL) fluid of Sftpc(-/-) mice. Viral titers and viral F and G protein mRNA were significantly increased in both Sftpc(-/-) and heterozygous Sftpc(+/-) mice compared with controls. Expression of Toll-like receptor 3 (TLR3) mRNA was increased in the lungs of Sftpc(-/-) mice relative to Sftpc(+/+) mice before and after RSV infection. Consistent with the increased TLR3 expression, BAL inflammatory cells were increased in the Sftpc(-/-) mice after exposure to a TLR3-specific ligand, poly(I:C). Preparations of purified SP-C and synthetic phospholipids blocked poly(I:C)-induced TLR3 signaling in vitro. SP-C deficiency increases the severity of RSV-induced pulmonary inflammation through regulation of TLR3 signaling.


Assuntos
Proteína C Associada a Surfactante Pulmonar/deficiência , Infecções por Vírus Respiratório Sincicial/metabolismo , Infecções por Vírus Respiratório Sincicial/patologia , Animais , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/virologia , Contagem de Células , Linhagem Celular , Colectinas/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Regulação Viral da Expressão Gênica , Células Caliciformes/patologia , Células Caliciformes/virologia , Humanos , Hipertrofia , Ligantes , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Camundongos , Pneumonia/complicações , Pneumonia/patologia , Pneumonia/virologia , Proteína C Associada a Surfactante Pulmonar/metabolismo , RNA de Cadeia Dupla/metabolismo , Infecções por Vírus Respiratório Sincicial/complicações , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/genética , Fatores de Tempo , Receptor 3 Toll-Like/metabolismo
20.
Am J Respir Cell Mol Biol ; 41(5): 562-72, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19244201

RESUMO

Transforming growth factor (TGF)-alpha is a ligand for the epidermal growth factor receptor (EGFR). EGFR activation is associated with fibroproliferative processes in human lung disease and animal models of pulmonary fibrosis. Overexpression of TGF-alpha in transgenic mice causes progressive and severe pulmonary fibrosis; however, the intracellular signaling pathways downstream of EGFR mediating this response are unknown. Using a doxycycline-regulatable transgenic mouse model of lung-specific TGF-alpha expression, we observed increased PCNA protein and phosphorylation of Akt and p70S6K in whole lung homogenates in association with induction of TGF-alpha. Induction in the lung of TGF-alpha caused progressive pulmonary fibrosis over a 7-week period. Daily administration of rapamycin prevented accumulation of total lung collagen, weight loss, and changes in pulmonary mechanics. Treatment of mice with rapamycin 4 weeks after the induction of TGF-alpha prevented additional weight loss, increases in total collagen, and changes in pulmonary mechanics. Rapamycin prevented further increases in established pulmonary fibrosis induced by EGFR activation. This study demonstrates that mammalian target of rapamycin (mTOR) is a major effector of EGFR-induced pulmonary fibrosis, providing support for further studies to determine the role of mTOR in the pathogenesis and treatment of pulmonary fibrosis.


Assuntos
Proteínas de Transporte/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Pulmão/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fibrose Pulmonar/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Fator de Crescimento Transformador alfa/metabolismo , Animais , Proteínas de Transporte/metabolismo , Colágeno/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Doxiciclina/farmacologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Cloridrato de Erlotinib , Regulação da Expressão Gênica , Humanos , Pulmão/enzimologia , Pulmão/fisiopatologia , Camundongos , Camundongos Transgênicos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fibrose Pulmonar/genética , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/fisiopatologia , Quinazolinas/farmacologia , Mecânica Respiratória/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Serina-Treonina Quinases TOR , Fatores de Tempo , Fator de Crescimento Transformador alfa/genética , Uteroglobina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...