Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Lung Cell Mol Physiol ; 309(8): L879-87, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26254422

RESUMO

Pulmonary fibrosis is a common and dose-limiting side-effect of ionizing radiation used to treat cancers of the thoracic region. Few effective therapies are available for this disease. Pulmonary fibrosis is characterized by an accumulation of myofibroblasts and excess deposition of extracellular matrix proteins. Although prior studies have reported that ionizing radiation induces fibroblast to myofibroblast differentiation and collagen production, the mechanism remains unclear. Transforming growth factor-ß (TGF-ß) is a key profibrotic cytokine that drives myofibroblast differentiation and extracellular matrix production. However, its activation and precise role in radiation-induced fibrosis are poorly understood. Recently, we reported that lactate activates latent TGF-ß through a pH-dependent mechanism. Here, we wanted to test the hypothesis that ionizing radiation leads to excessive lactate production via expression of the enzyme lactate dehydrogenase-A (LDHA) to promote myofibroblast differentiation. We found that LDHA expression is increased in human and animal lung tissue exposed to ionizing radiation. We demonstrate that ionizing radiation induces LDHA, lactate production, and extracellular acidification in primary human lung fibroblasts in a dose-dependent manner. We also demonstrate that genetic and pharmacologic inhibition of LDHA protects against radiation-induced myofibroblast differentiation. Furthermore, LDHA inhibition protects from radiation-induced activation of TGF-ß. We propose a profibrotic feed forward loop, in which radiation induces LDHA expression and lactate production, which can lead to further activation of TGF-ß to drive the fibrotic process. These studies support the concept of LDHA as an important therapeutic target in radiation-induced pulmonary fibrosis.


Assuntos
L-Lactato Desidrogenase/metabolismo , Miofibroblastos/efeitos da radiação , Animais , Diferenciação Celular/efeitos da radiação , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Gossipol/farmacologia , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , L-Lactato Desidrogenase/antagonistas & inibidores , Lactato Desidrogenase 5 , Ácido Láctico/biossíntese , Pulmão/enzimologia , Pulmão/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Miofibroblastos/citologia , Miofibroblastos/enzimologia , Fibrose Pulmonar/enzimologia , Fibrose Pulmonar/etiologia , Lesões por Radiação/enzimologia , Lesões por Radiação/etiologia , Fator de Crescimento Transformador beta/metabolismo
2.
Exp Eye Res ; 94(1): 136-45, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22178289

RESUMO

A critical component of corneal scarring is the TGFß-induced differentiation of corneal keratocytes into myofibroblasts. Inhibitors of this differentiation are potentially therapeutic for corneal scarring. In this study, we tested the relative effectiveness and mechanisms of action of two electrophilic peroxisome proliferator-activated receptor gamma (PPARγ) ligands: cyano-3,12-dioxolean-1,9-dien-28-oic acid-methyl ester (CDDO-Me) and 15-deoxy-Δ(-12,14)-prostaglandin J(2) (15d-PGJ(2)) for inhibiting TGFß-induced myofibroblast differentiation in vitro. TGFß was used to induce myofibroblast differentiation in cultured, primary human corneal fibroblasts. CDDO-Me and 15d-PGJ(2) were added to cultures to test their ability to inhibit this process. Myofibroblast differentiation was assessed by measuring the expression of myofibroblast-specific proteins (αSMA, collagen I, and fibronectin) and mRNA (αSMA and collagen III). The role of PPARγ in the inhibition of myofibroblast differentiation by these agents was tested in genetically and pharmacologically manipulated cells. Finally, we assayed the importance of electrophilicity in the actions of these agents on TGFß-induced αSMA expression via Western blotting and immunofluorescence. Both electrophilic PPARγ ligands (CDDO-Me and 15d-PGJ(2)) potently inhibited TGFß-induced myofibroblast differentiation, but PPARγ was only partially required for inhibition of myofibroblast differentiation by either agent. Electrophilic PPARγ ligands were able to inhibit myofibroblast differentiation more potently than non-electrophilic PPARγ ligands, suggesting an important role of electrophilicity in this process. CDDO-Me and 15d-PGJ(2) are strong inhibitors of TGFß-induced corneal fibroblast to myofibroblast differentiation in vitro, suggesting this class of agents as potential novel therapies for corneal scarring warranting further study in pre-clinical animal models.


Assuntos
Transdiferenciação Celular/efeitos dos fármacos , Córnea/citologia , Fibroblastos/citologia , Miofibroblastos/citologia , Ácido Oleanólico/análogos & derivados , PPAR gama/metabolismo , Prostaglandina D2/análogos & derivados , Actinas/genética , Actinas/metabolismo , Biomarcadores/metabolismo , Western Blotting , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Córnea/metabolismo , Fibroblastos/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Citometria de Fluxo , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Ligantes , Miofibroblastos/metabolismo , Ácido Oleanólico/farmacologia , Prostaglandina D2/farmacologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta/farmacologia
3.
Curr Pharm Des ; 14(21): 2051-60, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18691115

RESUMO

There is much interest in the potential use of Cox-2 selective inhibitors in combination with other cancer therapeutics. Malignancies of hematopoietic and non-hematopoietic origin often have increased expression of cyclooxygenase-2 (Cox-2), a key modulator of inflammation. For example, hematological malignancies such as chronic lymphocytic leukemia, chronic myeloid leukemia, Hodgkin's lymphoma, non-Hodgkin's lymphoma and multiple myeloma often highly express Cox-2, which correlates with poor patient prognosis. Expression of Cox-2 enhances survival and proliferation of malignant cells, while negatively influencing anti-tumor immunity. Hematological malignancies expressing elevated levels of Cox-2 potentially avoid immune responses by producing factors that enhance angiogenesis and metastasis. Cellular immune responses regulated by natural killer cells, cytotoxic T lymphocytes, and T regulatory cells are also influenced by Cox-2 expression. Therefore, Cox-2 selective inhibitors have promising therapeutic potential in patients suffering from certain hematological malignancies.


Assuntos
Inibidores de Ciclo-Oxigenase 2/farmacologia , Ciclo-Oxigenase 2/efeitos dos fármacos , Neoplasias Hematológicas/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Sistemas de Liberação de Medicamentos , Regulação Neoplásica da Expressão Gênica , Neoplasias Hematológicas/fisiopatologia , Humanos
4.
Eur Respir J ; 28(6): 1276-9, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17138682

RESUMO

Respiratory involvement is a frequent complication of Stevens-Johnson syndrome (SJS). However, there are very few convincing reports of persistent pulmonary sequelae, as demonstrated by spirometry, radiology and pathology. The current study presents a case of a 13-yr-old female with T-cell acute lymphocytic leukaemia who developed persistent, severe, obstructive lung disease following an episode of SJS. A lung biopsy demonstrated bronchiolar submucosal fibrosis consistent with constrictive bronchiolitis, as well as eosinophilic micro-abscesses, which, to the current authors' knowledge, has not been previously described. The present study illustrates specific histopathological features that highlight a possible association between Stevens-Johnson syndrome, constrictive bronchiolitis and eosinophilic micro-abscesses. The eosinophils may be associated with permanent mucosal damage, as seen in the present case, by releasing mediators that have a pro-fibrogenetic role. However, further investigation is warranted.


Assuntos
Obstrução das Vias Respiratórias/etiologia , Eosinofilia Pulmonar/etiologia , Síndrome de Stevens-Johnson/complicações , Adolescente , Obstrução das Vias Respiratórias/tratamento farmacológico , Obstrução das Vias Respiratórias/patologia , Feminino , Humanos , Eosinofilia Pulmonar/tratamento farmacológico , Eosinofilia Pulmonar/patologia , Testes de Função Respiratória , Síndrome de Stevens-Johnson/terapia
5.
Am J Physiol Lung Cell Mol Physiol ; 289(3): L391-9, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15863442

RESUMO

Cigarette smoking can lead to chronic lung inflammation and lung cancer. Chronic inflammation, associated with expression of cyclooxygenase-2 (COX-2) and prostaglandins, predisposes to malignancy. We recently demonstrated that human lung fibroblasts are activated by cigarette smoke to express COX-2 and prostaglandin E(2) (PGE(2)). Little is known about the mechanism whereby smoke activates human lung fibroblasts to produce proinflammatory mediators. Herein, we report the central role of the aryl hydrocarbon receptor (AHR) in cigarette smoke extract (CSE)-induced COX-2, microsomal PGE(2) synthase (mPGES), and PGE(2) production in human lung fibroblasts. Western blot analysis revealed that primary strains of human lung fibroblasts express AHR and aryl hydrocarbon nuclear translocator protein, supporting the possibility that smoke activates lung fibroblasts through this pathway. Experiments were subsequently performed to determine whether the AHR was activated by CSE. Immunocytochemistry and EMSA analysis revealed that CSE induced nuclear translocation of the AHR in human lung fibroblasts. CSE decreased protein levels of the AHR, consistent with AHR ligand-induced proteosome-mediated degradation. CSE also induced mPGES-1 and COX-2 protein and increased PGE(2) production. Treatment of human fibroblasts with AHR antagonists in the presence of CSE inhibited AHR nuclear translocation as well as COX-2, mPGES-1, and PGE(2) production. These data indicate that the AHR pathway plays an important role in cigarette smoke-mediated COX-2 and PG production in human lung fibroblasts and may contribute to tobacco-associated inflammation and lung disease.


Assuntos
Dinoprostona/biossíntese , Pulmão/metabolismo , Nicotiana , Prostaglandina-Endoperóxido Sintases/metabolismo , Receptores de Hidrocarboneto Arílico/fisiologia , Fumaça , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Translocador Nuclear Receptor Aril Hidrocarboneto , Células Cultivadas , Ciclo-Oxigenase 2 , Proteínas de Ligação a DNA/metabolismo , Fibroblastos/metabolismo , Flavonas/farmacologia , Flavonoides/farmacologia , Humanos , Oxirredutases Intramoleculares/biossíntese , Pulmão/citologia , Proteínas de Membrana , Prostaglandina-E Sintases , Receptores de Hidrocarboneto Arílico/antagonistas & inibidores , Receptores de Hidrocarboneto Arílico/metabolismo , Fatores de Transcrição/metabolismo
6.
Am J Physiol Lung Cell Mol Physiol ; 289(2): L322-8, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15833762

RESUMO

It has been hypothesized that the destruction of lung tissue observed in smokers with chronic obstructive pulmonary disease and emphysema is mediated by neutrophils recruited to the lungs by smoke exposure. This study investigated the role of the chemokine receptor CXCR2 in mediating neutrophilic inflammation in the lungs of mice acutely exposed to cigarette smoke. Exposure to dilute mainstream cigarette smoke for 1 h, twice per day for 3 days, induced acute inflammation in the lungs of C57BL/6 mice, with increased neutrophils and the neutrophil chemotactic CXC chemokines macrophage inflammatory protein (MIP)-2 and KC. Treatment with SCH-N, an orally active small molecule inhibitor of CXCR2, reduced the influx of neutrophils into the bronchoalveolar lavage (BAL) fluid. Histological changes were seen, with drug treatment reducing perivascular inflammation and the number of tissue neutrophils. beta-Glucuronidase activity was reduced in the BAL fluid of mice treated with SCH-N, indicating that the reduction in neutrophils was associated with a reduction in tissue damaging enzymes. Interestingly, whereas MIP-2 and KC were significantly elevated in the BAL fluid of smoke exposed mice, they were further elevated in mice exposed to smoke and treated with drug. The increase in MIP-2 and KC with drug treatment may be due to the decrease in lung neutrophils that either are not present to bind these chemokines or fail to provide a feedback signal to other cells producing these chemokines. Overall, these results demonstrate that inhibiting CXCR2 reduces neutrophilic inflammation and associated lung tissue damage due to acute cigarette smoke exposure.


Assuntos
Pulmão/efeitos dos fármacos , Nicotiana/toxicidade , Pneumonia/metabolismo , Receptores de Interleucina-8B/metabolismo , Fumaça/efeitos adversos , Doença Aguda , Animais , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Quimiocina CXCL2 , Feminino , Glucuronidase/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Monocinas/metabolismo , Neutrófilos/metabolismo , Neutrófilos/patologia , Pneumonia/induzido quimicamente
7.
Eur Respir J ; 23(6): 954-6, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15219012

RESUMO

A case of pulmonary hyalinising granuloma (PHG) complicated by deep venous thrombosis (DVT) is presented. The DVT was associated with the presence of a lupus anticoagulant. In the past PHG has been linked to various auto-antibodies, but to the best of the authors' knowledge, this is the first case reporting PHG in association with a lupus anticoagulant and clinically significant venous thrombosis. Historically, PHG has been regarded as poorly corticosteroid responsive. However, the patient in this case study responded dramatically to prednisone. This case study suggests that in selected patients with pulmonary hyalinising granuloma experiencing disabling symptoms and worsening pulmonary function, a trial of corticosteroids may be warranted.


Assuntos
Granuloma do Sistema Respiratório/complicações , Pneumopatias/complicações , Trombose Venosa/complicações , Anticoagulantes/uso terapêutico , Granuloma do Sistema Respiratório/patologia , Humanos , Pneumopatias/patologia , Inibidor de Coagulação do Lúpus/imunologia , Masculino , Pessoa de Meia-Idade , Trombose Venosa/tratamento farmacológico , Trombose Venosa/imunologia
8.
Am J Respir Cell Mol Biol ; 25(1): 3-7, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11472967

RESUMO

Tumor necrosis factor-alpha receptor knockout (TNF-alphaRKO) mice have homozygous deletions of the genes that code for both the 55- and 75-kD receptors. The mice are protected from the fibrogenic effects of bleomycin, silica, and inhaled asbestos. The asbestos-exposed animals exhibit reduced expression of other peptide growth factors such as transforming growth factor (TGF)-alpha, platelet-derived growth factors, and TGF-beta. In normal animals, these and other cytokines are elaborated at high levels during the development of fibroproliferative lung disease, but there is little information available that has allowed investigators to establish the role of the individual growth factors in disease pathogenesis. Here, we show that overexpression of TGF-beta(1) by means of a replication-deficient adenovirus vector induces fibrogenesis in the lungs of the fibrogenic-resistant TNF-alphaRKO mice. The fibrogenic lesions developed in both the KO and background controls within 7 d, and both types of animals exhibited similar incorporation of bromodeoxyuridine. Interestingly, airway epithelial cell proliferation appeared to be suppressed, perhaps due to the presence of the TGF-beta(1), a well-known inhibitor of epithelial mitogenesis. Before these experiments, there was no information available that would provide a basis for predicting whether or not TGF-beta(1) expression induces fibroproliferative lung disease in fibrogenic-resistant TNF-alphaRKO mice, an increasingly popular animal model.


Assuntos
Fibrose Pulmonar/genética , Receptores do Fator de Necrose Tumoral/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Adenoviridae/genética , Animais , Bromodesoxiuridina , Vetores Genéticos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores do Fator de Necrose Tumoral/genética , Transdução Genética , Fator de Crescimento Transformador beta/genética
9.
Clin Immunol ; 99(3): 308-19, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11358425

RESUMO

Tissue fibrosis can lead to significant organ dysfunction and resulting patient morbidity and mortality. Unfortunately, the therapeutic repertoire is currently limited, nonspecific, and largely ineffective. While the pathogenesis is incompletely understood, evidence is accumulating that immune and cytokine mediated mechanisms are critical. In this review, data will be provided to support the role of Type 2 cytokines in the pathogenesis of fibrosis. The importance of the role of the pro-fibrogenic cytokine TGF-beta and CD40-CD40 ligand mediated fibroblast activation will also be evaluated. Finally, novel therapeutic options based on inhibiting these pathways will be described.


Assuntos
Fibrose/etiologia , Fibrose Pulmonar/etiologia , Animais , Antígenos CD40/fisiologia , Ligante de CD40/fisiologia , Citocinas/fisiologia , Fibrose/terapia , Humanos , Interferon gama/uso terapêutico , Fibrose Pulmonar/terapia , Fator de Crescimento Transformador beta/fisiologia
10.
Am J Physiol Lung Cell Mol Physiol ; 280(6): L1327-34, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11350814

RESUMO

Transforming growth factor (TGF)-beta is a key cytokine in the pathogenesis of pulmonary fibrosis, and pharmacological interference with TGF-beta can ameliorate the fibrotic tissue response. The small proteoglycans decorin and biglycan are able to bind and inhibit TGF-beta activity in vitro. Although decorin has anti-TGF-beta properties in vivo, little is known about the physiological role of biglycan in vivo. Adenoviral gene transfer was used to overexpress active TGF-beta, decorin, and biglycan in cell culture and in murine lungs. Both proteoglycans were able to interfere with TGF-beta bioactivity in vitro in a dose-dependant manner. In vivo, overexpression of TGF-beta resulted in marked lung fibrosis, which was significantly reduced by concomitant overexpression of decorin. Biglycan, however, had no significant effect on lung fibrosis induced by TGF-beta. The data suggest that differences in tissue distribution are responsible for the different effects on TGF-beta bioactivity in vivo, indicating that decorin, but not biglycan, has potential therapeutic value in fibrotic disorders of the lung.


Assuntos
Pulmão/metabolismo , Proteoglicanas/farmacologia , Fibrose Pulmonar/prevenção & controle , Fator de Crescimento Transformador beta/antagonistas & inibidores , Adenoviridae/genética , Animais , Biglicano , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Linhagem Celular , Decorina , Relação Dose-Resposta a Droga , Proteínas da Matriz Extracelular , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Vetores Genéticos/farmacologia , Humanos , Hidroxiprolina/metabolismo , Imuno-Histoquímica , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Proteoglicanas/genética , Proteoglicanas/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Distribuição Tecidual , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/genética , Resultado do Tratamento
11.
Am J Respir Crit Care Med ; 163(3 Pt 1): 770-7, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11254537

RESUMO

Pulmonary fibrosis is a chronic progressive disease with no effective therapy. Transforming growth factor beta (TGF-beta) is thought to be a key profibrotic mediator and blocking its activity is therefore one of the targets of new treatment strategies for fibrosis. Decorin is an endogenous proteoglycan and one of the known inhibitors of TGF-beta. The short half-life of peptide-based therapeutics makes gene transfer a promising approach to achieve prolonged protein levels in the lung. Replication-deficient adenovirus was used to deliver decorin transgene (AdDec) to the airways by a single intranasal injection in a murine bleomycin model of lung fibrosis. The ability of vector-derived decorin to inhibit TGF-beta was examined in a bioassay and its effect on bleomycin-induced pulmonary fibrosis was determined by histomorphology and lung hydroxyproline. In vitro, supernatant from cells infected with AdDec abrogated the bioactivity of TGF-beta in a dose-dependent manner whereas control virus (AdDL70) had no effect. In vivo, treatment of bleomycin-injected mice with AdDec substantially reduced the fibrogenic response compared with control virus (hydroxyproline: bleomycin/AdDec, 1.96 microg/mg; bleomycin/AdDL70, 3.05 microg/mg; p = 0.0005). These results suggest that a single administration of AdDec was able to generate a local pulmonary environment that effectively blocked the fibrogenic response to bleomycin by inhibition of TGF-beta.


Assuntos
Antibióticos Antineoplásicos , Bleomicina , Proteoglicanas/genética , Fibrose Pulmonar/genética , Fator de Crescimento Transformador beta/antagonistas & inibidores , Transgenes/genética , Adenoviridae , Animais , Antibióticos Antineoplásicos/administração & dosagem , Bleomicina/administração & dosagem , Decorina , Proteínas da Matriz Extracelular , Feminino , Hidroxiprolina/análise , Pulmão/química , Camundongos , Camundongos Endogâmicos C57BL , Fibrose Pulmonar/induzido quimicamente , Células Tumorais Cultivadas
12.
Eur J Cell Biol ; 78(10): 715-25, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10569244

RESUMO

The precise spatial-temporal role that expression and activation of transforming growth factor (TGF)-beta plays in mammalian organ morphogenesis remains incompletely understood. Using replication deficient adenoviral vectors containing engineered TGF-beta1 cDNAs, we studied the spatial effects of locally over-expressing either latent or mutated, constitutively active TGF-beta1 protein during embryonic mouse lung branching morphogenesis in culture. Transfer of exogenous genes into lung epithelium was achieved by intra-tracheal micro-injection of recombinant adenovirus, while submerging lungs in virus resulted in gene transfer into the pleura and subjacent mesenchymal cells, as revealed by cytochemical staining for beta-galactosidase. Only lungs transfected with active, but not latent TGF-beta1 gene, showed elevated levels of active TGF-beta. Epithelial over-expression of active, but not latent TGF-beta1, via intra-tracheal micro-injection inhibited lung branching morphogenesis by 36 %. In contrast, lungs submerged with either active or latent TGF-beta1 recombinant virus did not demonstrate an inhibitory effect upon branching. Pulmonary gene regulation was assayed by competitive polymerase chain reaction coupled with reverse transcription. Direct respiratory tract micro-injection of adenovirus over-expressing active TGF-beta1 resulted in a dose-dependent inhibition of epithelial surfactant protein (SP)-C and SP-B mRNA levels by up to 76 % and 70 %, respectively, while in contrast, fibronectin and matrix Gla protein (MGP) mRNA levels remained stable. However, lungs that had been submerged in adenovirus expressing active TGF-beta1 demonstrated a concentration-dependent induction of both fibronectin and MGP mRNA levels up to 4.3- and 4.7-fold respectively in the presence of 1 x 10(11) pfu/ml active TGF-beta1 virus. On the other hand, lungs treated with adenovirus expressing latent TGF-beta1 either by micro-injection or submerging failed to demonstrate any regulatory effect either upon epithelial or mesenchymal gene expression. We conclude that adenovector-mediated over-expression of activated TGF-beta1 in specific spatial compartments results respectively in either inhibition of branching morphogenesis and epithelium-specific gene expression, or in induction of matrix gene expression without affecting morphogenesis or epithelium-specific gene expression, depending on the route of administration. Also, the lack of effect of latent TGF-beta1 over-expression strongly suggests that TGF-beta activation per se provides an important locus of fine regulation of the spatial effects of TGF-beta signaling during embryonic lung branching morphogenesis.


Assuntos
Pulmão/embriologia , Pulmão/metabolismo , Fator de Crescimento Transformador beta/genética , Adenoviridae/genética , Animais , Sequência de Bases , Primers do DNA/genética , Proteínas da Matriz Extracelular/genética , Fibronectinas/genética , Regulação da Expressão Gênica no Desenvolvimento , Vetores Genéticos , Camundongos , Morfogênese , Mutação , Técnicas de Cultura de Órgãos , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transfecção
13.
Am J Physiol ; 277(2): L412-22, 1999 08.
Artigo em Inglês | MEDLINE | ID: mdl-10444536

RESUMO

Excessive transforming growth factor (TGF)-beta signaling has been implicated in pulmonary hypoplasia associated with bronchopulmonary dysplasia, a chronic lung disease of human prematurity featuring pulmonary fibrosis. This implies that inhibitors of TGF-beta could be useful therapeutic agents. Because exogenous TGF-beta ligands are known to inhibit lung branching morphogenesis and cytodifferentiation in mouse embryonic lungs in ex vivo culture, we examined the capacity of a naturally occurring inhibitor of TGF-beta activity, the proteoglycan decorin, to overcome the inhibitory effects of exogenous TGF-beta. Intratracheal microinjection of a recombinant adenovirus containing decorin cDNA resulted in overexpression of the exogenous decorin gene in airway epithelium. Although exogenous TGF-beta efficiently decreased epithelial lung branching morphogenesis in control cultures, TGF-beta-induced inhibition of lung growth was abolished after epithelial transfer of the decorin gene. Additionally, exogenous TGF-beta-induced antiproliferative effects as well as the downregulation of surfactant protein C were abrogated by decorin in cultured embryonic lungs. Moreover, lung branching inhibition by TGF-beta could be restored by the addition of decorin antisense oligodeoxynucleotides in culture, indicating that decorin is both specifically and directly involved in suppressing TGF-beta-mediated negative regulation of lung morphogenesis. Our findings suggest that decorin can antagonize bioactive TGF-beta during lung growth and differentiation, establishing the rationale for decorin as a candidate therapeutic approach to ameliorate excessive levels of TGF-beta signaling in the developing lung.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Pulmão/embriologia , Proteoglicanas/genética , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Ciclina A/genética , Decorina , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/fisiologia , Desenvolvimento Embrionário e Fetal/efeitos dos fármacos , Desenvolvimento Embrionário e Fetal/fisiologia , Epitélio/embriologia , Proteínas da Matriz Extracelular , Expressão Gênica/efeitos dos fármacos , Vetores Genéticos , Camundongos , Microinjeções , Oligonucleotídeos Antissenso/farmacologia , Técnicas de Cultura de Órgãos , Proteoglicanas/metabolismo , Proteolipídeos/genética , Surfactantes Pulmonares/genética , Traqueia , Fator de Crescimento Transformador beta/farmacologia
16.
Am J Pathol ; 153(3): 825-32, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9736031

RESUMO

Tumor necrosis factor-alpha is up-regulated in a variety of different human immune-inflammatory and fibrotic pulmonary pathologies. However, its precise role in these pathologies and, in particular, the mechanism(s) by which it may induce fibrogenesis are not yet elucidated. Using a replication-deficient adenovirus to transfer the cDNA of tumor necrosis factor-alpha to rat lung, we have been able to study the effect of transient but prolonged (7 to 10 days) overexpression of tumor necrosis factor-alpha in normal adult pulmonary tissue. We have demonstrated that local overexpression resulted in severe pulmonary inflammation with significant increases in neutrophils, macrophages, and lymphocytes and, to a lesser extent, eosinophils, with a peak at day 7. By day 14, the inflammatory cell accumulation had declined, and fibrogenesis became evident, with fibroblast accumulation and deposition of extracellular matrix proteins. Fibrotic changes were patchy but persisted to beyond day 64. To elucidate the mechanism underlying this fibrogenesis, we examined bronchoalveolar fluids for the presence of the fibrogenic cytokine transforming growth factor-beta1 and tissues for induction of alpha-smooth muscle actin-rich myofibroblasts. Transforming growth factor-beta1 was transiently elevated from day 7 (peak at day 14) immediately preceding the onset of fibrogenesis. Furthermore, there was a striking accumulation of myofibroblasts from day 7, with the most extensive and intense immunostaining at day 14, ie, coincident with the up-regulation of transforming growth factor-beta1 and onset of fibrogenesis. Thus, we have provided a model of tumor necrosis factor-alpha-mediated pulmonary inflammation and fibrosis in normal adult lung, and we suggest that the fibrogenesis may be mediated by the secondary up-regulation of transforming growth factor-beta1 and induction of pulmonary myofibroblasts.


Assuntos
Técnicas de Transferência de Genes , Pulmão/metabolismo , Pneumonia/etiologia , Fibrose Pulmonar/etiologia , Fator de Crescimento Transformador beta/biossíntese , Fator de Necrose Tumoral alfa/genética , Adenoviridae/genética , Animais , Líquido da Lavagem Broncoalveolar/citologia , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Expressão Gênica , Vetores Genéticos , Técnicas Imunoenzimáticas , Pulmão/patologia , Pulmão/virologia , Pneumonia/metabolismo , Pneumonia/patologia , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
17.
Mech Dev ; 72(1-2): 89-100, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9533955

RESUMO

Although exogenous transforming growth factor-beta (TGF-beta) is known to inhibit branching morphogenesis in mouse embryonic lungs in culture, whether the principal negative function of endogenous TGF-beta signaling resides in lung epithelium or mesenchyme remains unresolved. A recombinant adenovirus was constructed, containing a mutated human TGF-beta type II receptor with a truncated cytoplasmic kinase domain. We examined whether this dominant-negative receptor could abolish epithelium-specific endogenous TGF-beta signaling. We introduced the recombinant adenovirus into lung explants via intra-tracheal micro-injection. This resulted in over-expression of exogenous truncated TGF-beta type II receptor only in airway epithelium, not in mesenchyme, as assessed by mRNA level and protein localization. Blockade of endogenous TGF-beta receptor signaling in epithelial endoderm by the mutated dominant-negative TGF-beta type II receptor resulted in significant (65%) stimulation of epithelial branching morphogenesis, while exogenous TGF-beta no longer downregulated epithelial PCNA immunoreactivity and surfactant protein C (SP-C) expression. Additionally, the mitogenic responses to epidermal growth factor (EGF) and platelet-derived growth factor, PDGF-AA were potentiated by 33 and 31%, respectively. We conclude that epithelium-specific adenovirus-mediated over-expression of a dominant-negative TGF-beta type II receptor completely and specifically abolished the anti-proliferative effects of both endogenous and exogenous TGF-beta. Therefore, epithelium-specific TGF-beta signaling is sufficient to negatively regulate embryonic lung-branching morphogenesis in culture. We speculate that abrogation of TGF-beta signaling stimulates lung morphogenesis by potentiating the inductive and permissive effects of other endogenous peptide growth factors such as EGF and PDGF-AA.


Assuntos
Fator de Crescimento Epidérmico/fisiologia , Células Epiteliais/fisiologia , Pulmão/embriologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Adenoviridae , Animais , Técnicas de Cultura , Densitometria , Feminino , Humanos , Camundongos , Morfogênese , Gravidez , Proteínas Serina-Treonina Quinases/fisiologia , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Fator de Crescimento Transformador beta/fisiologia
18.
Clin Exp Allergy ; 28(12): 1581-90, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10024231

RESUMO

BACKGROUND: Recent epidemiological studies have suggested that exposure to certain viruses and bacteria influences the development of allergy and allergic diseases, such as asthma. However, there is a paucity of experimental evidence examining the consequences of concurrent exposure to allergen and infectious agents, and the potential mechanisms by which allergic disease might be averted as a result. OBJECTIVE: To model this situation experimentally, we investigated whether a virally induced immune response, elicited by a replication-deficient human type 5 adenovirus (RDA) administered at a site distant from the airways, could inhibit ovalbumin (OVA)-induced airways eosinophilic inflammation. METHODS: C57BL/6 mice were infected intramuscularly with RDA 16h prior to intraperitoneal OVA sensitization. Cellular and cytokine responses in the lung/airways were examined after an OVA aerosol challenge. RESULTS: RDA infection significantly inhibited the inflammatory response in the lung tissue after antigen challenge. In the bronchoalveolar lavage (BAL), total cell number, eosinophils and lymphocytes were decreased by 70, 85 and 65%, respectively, after antigen challenge in RDA-treated, compared with untreated, mice. RDA infection had no effect on IgE synthesis. The levels of IL-5, IL-4 and IFNgamma in the BAL after antigen challenge were significantly lower in RDA-treated mice. In vitro production of cytokines by splenocytes in response to OVA restimulation revealed a shift from IL-4 in sensitized, PBS-treated mice, to IFNgamma in sensitized mice treated with RDA. Flow cytometric analysis revealed that RDA infection increased the proportion of CD8 T cells in the BAL; this change in T-cell subsets was accompanied by an increase in both CD4 and CD8 T cells positive for intracellular IFNgamma. Inhibition of antigen-induced airways inflammation was IFNgamma-dependent but did not require IL-12, as RDA-treatment inhibited airways inflammation in IL-12 but not IFNgamma knock-out mice. CONCLUSION: This study demonstrates that an immune response against a replication-deficient adenovirus during the initial exposure to OVA inhibits the development of airways inflammation after antigen aerosol challenge.


Assuntos
Infecções por Adenoviridae/imunologia , Adenovírus Humanos/imunologia , Vírus Defeituosos/imunologia , Eosinófilos/imunologia , Pulmão/imunologia , Hipersensibilidade Respiratória/imunologia , Animais , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/biossíntese , Feminino , Citometria de Fluxo , Humanos , Imunoglobulina E/biossíntese , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Hipersensibilidade Respiratória/patologia , Baço/citologia , Baço/imunologia , Subpopulações de Linfócitos T/imunologia , Replicação Viral
19.
J Clin Invest ; 100(4): 768-76, 1997 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-9259574

RESUMO

Transforming growth factor (TGF)-beta1 has been implicated in the pathogenesis of fibrosis based upon its matrix-inducing effects on stromal cells in vitro, and studies demonstrating increased expression of total TGF-beta1 in fibrotic tissues from a variety of organs. The precise role in vivo of this cytokine in both its latent and active forms, however, remains unclear. Using replication-deficient adenovirus vectors to transfer the cDNA of porcine TGF-beta1 to rat lung, we have been able to study the effect of TGF-beta1 protein in the respiratory tract directly. We have demonstrated that transient overexpression of active, but not latent, TGF-beta1 resulted in prolonged and severe interstitial and pleural fibrosis characterized by extensive deposition of the extracellular matrix (ECM) proteins collagen, fibronectin, and elastin, and by emergence of cells with the myofibroblast phenotype. These results illustrate the role of TGF-beta1 and the importance of its activation in the pulmonary fibrotic process, and suggest that targeting active TGF-beta1 and steps involved in TGF-beta1 activation are likely to be valuable antifibrogenic therapeutic strategies. This new and versatile model of pulmonary fibrosis can be used to study such therapies.


Assuntos
Fragmentos de Peptídeos , Precursores de Proteínas , Fibrose Pulmonar/etiologia , Fator de Crescimento Transformador beta/fisiologia , Actinas/análise , Adenoviridae/genética , Animais , Colágeno/análise , Elastina/análise , Fibronectinas/análise , Técnicas de Transferência de Genes , Vetores Genéticos , Hidroxiprolina/análise , Pulmão/metabolismo , Pulmão/patologia , Masculino , Proteínas/genética , Proteínas/metabolismo , Fibrose Pulmonar/patologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA