Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Biochimie ; 180: 90-103, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33122105

RESUMO

Gentisate 1,2-dioxygenases belong to the class III ring-cleaving dioxygenases catalyzing key reactions of aromatic compounds degradation by aerobic microorganisms. In the present work, the results of complete molecular, structural, and functional investigations of the gentisate 1,2-dioxygenase (rho-GDO) from a gram-positive bacterium Rhodococcus opacus 1CP growing on 3-hydroxybenzoate as a sole source of carbon and energy are presented. The purified enzyme showed a narrow substrate specificity. Among fourteen investigated substrate analogues only gentisate was oxidized by the enzyme, what can be potentially applied in biosensor technologies. The rho-GDO encoding gene was identified in the genomic DNA of the R. opacus 1CP. According to phylogenetic analysis, the rho-GDO belongs to the group of apparently most recently acquired activities in bacterial genera Rhodococcus, Arthrobacter, Corynebacterium, Nocardia, Amycolatopsis, Comamonas, and Streptomyces. Homology modeling the rho-GDO 3D-structure demonstrates the composition identity of the first-sphere residues of the active site of rho-GDO and salicylate 1,2-dioxygenase from Pseudaminobacter salicylatoxidans (RCSB PDB: 2PHD), despite of their different substrate specificities. The phenomenon described for the first time for this family of enzymes supposes a more complicated mechanism of substrate specificity than previously imagined, and makes the rho-GDO a convenient model for a novel direction of structure-function relationship studies.


Assuntos
Dioxigenases/química , Dioxigenases/metabolismo , Rhodococcus/enzimologia , Rhodococcus/genética , Domínio Catalítico , Clonagem Molecular , Dioxigenases/isolamento & purificação , Escherichia coli/genética , Cinética , Modelos Moleculares , Filogenia , Conformação Proteica , Rhodococcus/crescimento & desenvolvimento , Análise de Sequência de Proteína , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
2.
Genes (Basel) ; 11(1)2019 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-31877734

RESUMO

Strawberry (Fragaria) and raspberry (Rubus) are very popular crops, and improving their nutritional quality and disease resistance are important tasks in their breeding programs that are becoming increasingly based on use of functional DNA markers. We identified 118 microsatellite (simple sequence repeat-SSR) loci in the nucleotide sequences of flavonoid biosynthesis and pathogenesis-related genes and developed 24 SSR markers representing some of these structural and regulatory genes. These markers were used to assess the genetic diversity of 48 Fragaria and Rubus specimens, including wild species and rare cultivars, which differ in berry color, ploidy, and origin. We have demonstrated that a high proportion of the developed markers are transferable within and between Fragaria and Rubus genera and are polymorphic. Transferability and polymorphism of the SSR markers depended on location of their polymerase chain reaction (PCR) primer annealing sites and microsatellite loci in genes, respectively. High polymorphism of the SSR markers in regulatory flavonoid biosynthesis genes suggests their allelic variability that can be potentially associated with differences in flavonoid accumulation and composition. This set of SSR markers may be a useful molecular tool in strawberry and raspberry breeding programs for improvement anthocyanin related traits.


Assuntos
Flavonoides/biossíntese , Fragaria/genética , Repetições de Microssatélites , Rubus/genética , Vias Biossintéticas , Fragaria/classificação , Fragaria/metabolismo , Filogenia , Proteínas de Plantas/genética , Rubus/classificação , Rubus/metabolismo , Especificidade da Espécie
3.
Thromb Haemost ; 119(12): 1968-1980, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31705517

RESUMO

INTRODUCTION: Impaired plasminogen activation (PA) is causally related to the development of lung fibrosis. Prior studies demonstrate that enhanced PA in the lung limits the severity of scarring following injury and in vitro studies indicate that PA promotes matrix degradation and fibroblast apoptosis. These findings led us to hypothesize that increased PA in an in vivo model would enhance the resolution of established lung fibrosis in conjunction with increased myofibroblast apoptosis. METHODS: Transgenic C57BL/6 mice with doxycycline inducible lung-specific urokinase plasminogen activator (uPA) expression or littermate controls were treated (day 0) with bleomycin or saline. Doxycycline was initiated on days 1, 9, 14, or 21. Lung fibrosis, stiffness, apoptosis, epithelial barrier integrity, and inflammation were assessed. RESULTS: Protection from fibrosis with uPA upregulation from day 1 through day 28 was associated with reduced parenchymal stiffness as determined by atomic force microscopy. Initiation of uPA expression beginning in the late inflammatory or the early fibrotic phase reduced stiffness and fibrosis at day 28. Induction of uPA activity in mice with established fibrosis decreased lung collagen and lung stiffness while increasing myofibroblast apoptosis. Upregulation of uPA did not alter lung inflammation but was associated with improved epithelial cell homeostasis. CONCLUSION: Restoring intrapulmonary PA activity diminishes lung fibrogenesis and enhances the resolution of established lung fibrosis. This PA-mediated resolution is associated with increased myofibroblast apoptosis and improved epithelial cell homeostasis. These studies support the potential capacity of the lung to resolve existing scar in murine models.


Assuntos
Regulação da Expressão Gênica , Pulmão/patologia , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Apoptose , Bleomicina/farmacologia , Colágeno/metabolismo , Doxiciclina/farmacologia , Fibroblastos/metabolismo , Genótipo , Homeostase , Hidroxiprolina/farmacologia , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
4.
Physiol Rep ; 6(12): e13753, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29952109

RESUMO

Fibrosis of the lung constitutes a major clinical challenge and novel therapies are required to alleviate the associated morbidity and mortality. Investigating the antifibrotic efficacy of drugs that are already in clinical practice offers an efficient strategy to identify new therapies. The phosphodiesterase 4 (PDE4) inhibitors, approved for the treatment of chronic obstructive pulmonary disease, harbor therapeutic potential for pulmonary fibrosis by augmenting the activity of endogenous antifibrotic mediators that signal through cyclic AMP. In this study, we tested the efficacy of several PDE4 inhibitors including a novel compound (Compound 1) in a murine model of lung fibrosis that results from a targeted type II alveolar epithelial cell injury. We also compared the antifibrotic activity of PDE4 inhibition to the two therapies that are FDA-approved for idiopathic pulmonary fibrosis (pirfenidone and nintedanib). We found that both preventative (day 0-21) and therapeutic (day 11-21) dosing regimens of the PDE4 inhibitors significantly ameliorated the weight loss and lung collagen accumulation that are the sequelae of targeted epithelial cell damage. In a therapeutic protocol, the reduction in lung fibrosis with PDE4 inhibitor administration was equivalent to pirfenidone and nintedanib. Treatment with this class of drugs also resulted in a decrease in plasma surfactant protein D concentration, a reduction in the plasma levels of several chemokines implicated in lung fibrosis, and an in vitro inhibition of fibroblast profibrotic gene expression. These results motivate further investigation of PDE4 inhibition as a treatment for patients with fibrotic lung disease.


Assuntos
Células Epiteliais Alveolares/patologia , Benzamidas/uso terapêutico , Isoquinolinas/uso terapêutico , Inibidores da Fosfodiesterase 4/uso terapêutico , Fibrose Pulmonar/tratamento farmacológico , Aminopiridinas/uso terapêutico , Animais , Benzamidas/administração & dosagem , Benzamidas/sangue , Células Cultivadas , Quimiocinas/sangue , AMP Cíclico/metabolismo , Ciclopropanos/uso terapêutico , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Fibroblastos/metabolismo , Humanos , Isoquinolinas/administração & dosagem , Isoquinolinas/sangue , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibidores da Fosfodiesterase 4/administração & dosagem , Inibidores da Fosfodiesterase 4/sangue , Fibrose Pulmonar/sangue , Fibrose Pulmonar/prevenção & controle , Proteína D Associada a Surfactante Pulmonar/sangue , Piridinas/uso terapêutico , Fator de Necrose Tumoral alfa/metabolismo
5.
Am J Respir Cell Mol Biol ; 54(4): 482-92, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26378893

RESUMO

Accumulation of apoptosis-resistant fibroblasts is a hallmark of pulmonary fibrosis. We hypothesized that disruption of inhibitor of apoptosis protein (IAP) family proteins would limit lung fibrosis. We first show that transforming growth factor-ß1 and bleomycin increase X-linked IAP (XIAP) and cellular IAP (cIAP)-1 and -2 in murine lungs and mesenchymal cells. Functional blockade of XIAP and the cIAPs with AT-406, an orally bioavailable second mitochondria-derived activator of caspases (Smac) mimetic, abrogated bleomycin-induced lung fibrosis when given both prophylactically and therapeutically. To determine whether the reduction in fibrosis was predominantly due to AT-406-mediated inhibition of XIAP, we compared the fibrotic response of XIAP-deficient mice (XIAP(-/y)) with littermate controls and found no difference. We found no alterations in total inflammatory cells of either wild-type mice treated with AT-406 or XIAP(-/y) mice. AT-406 treatment limited CCL12 and IFN-γ production, whereas XIAP(-/y) mice exhibited increased IL-1ß expression. Surprisingly, XIAP(-/y) mesenchymal cells had increased resistance to Fas-mediated apoptosis. Functional blockade of cIAPs with AT-406 restored sensitivity to Fas-mediated apoptosis in XIAP(-/y) mesenchymal cells in vitro and increased apoptosis of mesenchymal cells in vivo, indicating that the increased apoptosis resistance in XIAP(-/y) mesenchymal cells was the result of increased cIAP expression. Collectively, these results indicate that: (1) IAPs have a role in the pathogenesis of lung fibrosis; (2) a congenital deficiency of XIAP may be overcome by compensatory mechanisms of other IAPs; and (3) broad functional inhibition of IAPs may be an effective strategy for the treatment of lung fibrosis by promoting mesenchymal cell apoptosis.


Assuntos
Bleomicina/toxicidade , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Fibrose Pulmonar/prevenção & controle , Animais , Apoptose , Azocinas/farmacologia , Compostos Benzidrílicos/farmacologia , Proteínas Inibidoras de Apoptose/genética , Interferon gama/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quimioatraentes de Monócitos/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , RNA Mensageiro/metabolismo , Fator de Crescimento Transformador beta1/administração & dosagem
6.
Am J Pathol ; 185(4): 969-86, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25681733

RESUMO

Myofibroblasts are crucial to the pathogenesis of tissue fibrosis. Their formation of stress fibers results in the release of myocardin-related transcription factor (MRTF), a transcriptional coactivator of serum response factor (SRF). MRTF-A (Mkl1)-deficient mice are protected from lung fibrosis. We hypothesized that the SRF/MRTF pathway inhibitor CCG-203971 would modulate myofibroblast function in vitro and limit lung fibrosis in vivo. Normal and idiopathic pulmonary fibrosis lung fibroblasts were treated with/without CCG-203971 (N-[4-chlorophenyl]-1-[3-(2-furanyl)benzoyl]-3-piperidine carboxamide) and/or Fas-activating antibody in the presence/absence of transforming growth factor (TGF)-ß1, and apoptosis was assessed. In vivo studies examined the effect of therapeutically administered CCG-203971 on lung fibrosis in two distinct murine models of fibrosis induced by bleomycin or targeted type II alveolar epithelial injury. In vitro, CCG-203971 prevented nuclear localization of MRTF-A; increased the apoptotic susceptibility of normal and idiopathic pulmonary fibrosis fibroblasts; blocked TGF-ß1-induced myofibroblast differentiation; and inhibited TGF-ß1-induced expression of fibronectin, X-linked inhibitor of apoptosis, and plasminogen activator inhibitor-1. TGF-ß1 did not protect fibroblasts or myofibroblasts from apoptosis in the presence of CCG-203971. In vivo, CCG-203971 significantly reduced lung collagen content in both murine models while decreasing alveolar plasminogen activator inhibitor-1 and promoting myofibroblast apoptosis. These data support a central role of the SRF/MRTF pathway in the pathobiology of lung fibrosis and suggest that its inhibition can help resolve lung fibrosis by promoting fibroblast apoptosis.


Assuntos
Apoptose , Pulmão/metabolismo , Pulmão/patologia , Mesoderma/patologia , Fator de Resposta Sérica/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Adulto , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/patologia , Animais , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Citoproteção/efeitos dos fármacos , Fibronectinas/metabolismo , Fibrose , Humanos , Inflamação/patologia , Mesoderma/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Miofibroblastos/patologia , Ácidos Nipecóticos/administração & dosagem , Ácidos Nipecóticos/farmacologia , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Transporte Proteico/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sus scrofa , Fator de Crescimento Transformador beta1/farmacologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Receptor fas/metabolismo
7.
J Immunol ; 190(7): 3447-57, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23467934

RESUMO

The alveolar epithelium is characteristically abnormal in fibrotic lung disease, and we recently established a direct link between injury to the type II alveolar epithelial cell (AEC) and the accumulation of interstitial collagen. The mechanisms by which damage to the epithelium induces lung scarring remain poorly understood. It is particularly controversial whether an insult to the type II AEC initiates an inflammatory response that is required for the development of fibrosis. To explore whether local inflammation occurs following a targeted epithelial insult and contributes to lung fibrosis, we administered diphtheria toxin to transgenic mice with type II AEC-restricted expression of the diphtheria toxin receptor. We used immunophenotyping techniques and diphtheria toxin receptor-expressing, chemokine receptor-2-deficient (CCR2(-/-)) mice to determine the participation of lung leukocyte subsets in pulmonary fibrogenesis. Our results demonstrate that targeted type II AEC injury induces an inflammatory response that is enriched for CD11b(+) nonresident exudate macrophages (ExM) and their precursors, Ly-6C(high) monocytes. CCR2 deficiency abrogates the accumulation of both cell populations and protects mice from fibrosis, weight loss, and death. Further analyses revealed that the ExM are alternatively activated and that ExM and Ly-6C(high) monocytes express mRNA for IL-13, TGF-ß, and the collagen genes, COL1A1 and COLIIIA1. Furthermore, the accumulated ExM and Ly-6C(high) monocytes contain intracellular collagen, as detected by immunostaining. Together, these results implicate CCR2 and the accumulation of ExM and Ly-6C(high) monocytes as critical determinants of pulmonary fibrosis induced by selective type II AEC injury.


Assuntos
Exsudatos e Transudatos/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Fibrose Pulmonar/genética , Fibrose Pulmonar/imunologia , Receptores CCR2/genética , Células Epiteliais Alveolares/imunologia , Células Epiteliais Alveolares/metabolismo , Animais , Antígenos Ly/imunologia , Colágeno/biossíntese , Citocinas/genética , Citocinas/imunologia , Exsudatos e Transudatos/citologia , Expressão Gênica , Marcação de Genes , Imunofenotipagem , Ativação de Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Monócitos/metabolismo , Fenótipo , Pneumonia/genética , Pneumonia/imunologia , Pneumonia/patologia , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/metabolismo , Fibrose Pulmonar/mortalidade , Receptores CCR2/imunologia , Redução de Peso/genética , Redução de Peso/imunologia
8.
J Pathol ; 228(2): 170-80, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22262246

RESUMO

Fibrotic disorders of the lung are associated with perturbations in the plasminogen activation system. Specifically, plasminogen activator inhibitor-1 (PAI-1) expression is increased relative to the plasminogen activators. A direct role for this imbalance in modulating the severity of lung scarring following injury has been substantiated in the bleomycin model of pulmonary fibrosis. However, it remains unclear whether derangements in the plasminogen activation system contribute more generally to the pathogenesis of lung fibrosis beyond bleomycin injury. To answer this question, we employed an alternative model of lung scarring, in which type II alveolar epithelial cells (AECs) are specifically injured by administering diphtheria toxin (DT) to mice genetically engineered to express the human DT receptor (DTR) off the surfactant protein C promoter. This targeted AEC injury results in the diffuse accumulation of interstitial collagen. In the present study, we found that this targeted type II cell insult also increases PAI-1 expression in the alveolar compartment. We identified AECs and lung macrophages to be sources of PAI-1 production. To determine whether this elevated PAI-1 concentration was directly related to the severity of fibrosis, DTR(+) mice were crossed into a PAI-1-deficient background (DTR(+) : PAI-1(-/-) ). DT administration to DTR(+) : PAI-1(-/-) animals caused significantly less fibrosis than was measured in DTR(+) mice with intact PAI-1 production. PAI-1 deficiency also abrogated the accumulation of CD11b(+) exudate macrophages that were found to express PAI-1 and type-1 collagen. These observations substantiate the critical function of PAI-1 in pulmonary fibrosis pathogenesis and provide new insight into a potential mechanism by which this pro-fibrotic molecule influences collagen accumulation. Copyright © 2012 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Células Epiteliais Alveolares/metabolismo , Macrófagos/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Fibrose Pulmonar/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/patologia , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/patologia , Animais , Líquido da Lavagem Broncoalveolar/química , Colágeno Tipo I/metabolismo , Toxina Diftérica/toxicidade , Modelos Animais de Doenças , Exsudatos e Transudatos/citologia , Exsudatos e Transudatos/efeitos dos fármacos , Exsudatos e Transudatos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidor 1 de Ativador de Plasminogênio/análise , Venenos/toxicidade , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo
9.
Blood ; 118(8): 2313-21, 2011 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-21734232

RESUMO

Plasminogen activator inhibitor-1 (PAI-1) is increased in the lungs of patients with pulmonary fibrosis, and animal studies have shown that experimental manipulations of PAI-1 levels directly influence the extent of scarring that follows lung injury. PAI-1 has 2 known properties that could potentiate fibrosis, namely an antiprotease activity that inhibits the generation of plasmin, and a vitronectin-binding function that interferes with cell adhesion to this extracellular matrix protein. To determine the relative importance of each PAI-1 function in lung fibrogenesis, we administered mutant PAI-1 proteins that possessed either intact antiprotease or vitronectin-binding activity to bleomycin-injured mice genetically deficient in PAI-1. We found that the vitronectin-binding capacity of PAI-1 was the primary determinant required for its ability to exacerbate lung scarring induced by intratracheal bleomycin administration. The critical role of the vitronectin-binding function of PAI-1 in fibrosis was confirmed in the bleomycin model using mice genetically modified to express the mutant PAI-1 proteins. We conclude that the vitronectin-binding function of PAI-1 is necessary and sufficient in its ability to exacerbate fibrotic processes in the lung.


Assuntos
Fibrose Pulmonar/etiologia , Fibrose Pulmonar/metabolismo , Serpina E2/metabolismo , Vitronectina/metabolismo , Animais , Bleomicina/administração & dosagem , Bleomicina/toxicidade , Líquido da Lavagem Broncoalveolar/química , Colágeno/metabolismo , Modelos Animais de Doenças , Humanos , Hidroxiprolina/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ligação Proteica , Fibrose Pulmonar/patologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serpina E2/sangue , Serpina E2/deficiência , Serpina E2/genética , Vitronectina/sangue
10.
Am J Respir Crit Care Med ; 181(3): 254-63, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19850947

RESUMO

RATIONALE: Ineffective repair of a damaged alveolar epithelium has been postulated to cause pulmonary fibrosis. In support of this theory, epithelial cell abnormalities, including hyperplasia, apoptosis, and persistent denudation of the alveolar basement membrane, are found in the lungs of humans with idiopathic pulmonary fibrosis and in animal models of fibrotic lung disease. Furthermore, mutations in genes that affect regenerative capacity or that cause injury/apoptosis of type II alveolar epithelial cells have been identified in familial forms of pulmonary fibrosis. Although these findings are compelling, there are no studies that demonstrate a direct role for the alveolar epithelium or, more specifically, type II cells in the scarring process. OBJECTIVES: To determine if a targeted injury to type II cells would result in pulmonary fibrosis. METHODS: A transgenic mouse was generated to express the human diphtheria toxin receptor on type II alveolar epithelial cells. Diphtheria toxin was administered to these animals to specifically target the type II epithelium for injury. Lung fibrosis was assessed by histology and hydroxyproline measurement. MEASUREMENTS AND MAIN RESULTS: Transgenic mice treated with diphtheria toxin developed an approximately twofold increase in their lung hydroxyproline content on Days 21 and 28 after diphtheria toxin treatment. The fibrosis developed in conjunction with type II cell injury. Histological evaluation revealed diffuse collagen deposition with patchy areas of more confluent scarring and associated alveolar contraction. CONCLUSIONS: The development of lung fibrosis in the setting of type II cell injury in our model provides evidence for a causal link between the epithelial defects seen in idiopathic pulmonary fibrosis and the corresponding areas of scarring.


Assuntos
Células Epiteliais/patologia , Alvéolos Pulmonares/patologia , Fibrose Pulmonar/patologia , Mucosa Respiratória/patologia , Animais , Apoptose/genética , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Genótipo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Peptídeos/genética , Peptídeos/metabolismo , Alvéolos Pulmonares/metabolismo , Fibrose Pulmonar/genética , Fibrose Pulmonar/metabolismo , Proteína C Associada a Surfactante Pulmonar , RNA/genética , Mucosa Respiratória/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Am J Respir Cell Mol Biol ; 31(6): 672-8, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15308506

RESUMO

The pathogenesis of pulmonary fibrosis is thought to involve alveolar epithelial injury that, when successfully repaired, can limit subsequent scarring. The plasminogen system participates in this process with the balance between urokinase-type plasminogen activator (uPA) and plasminogen activator inhibitor-1 (PAI-1) being a critical determinant of the extent of collagen accumulation that follows lung injury. Because the plasminogen system is known to influence the rate of migration of epithelial cells, including keratinocytes and bronchial epithelial cells, we hypothesized that the balance of uPA and PAI-1 would affect the efficiency of alveolar epithelial cell (AEC) wound repair. Using an in vitro model of AEC wounding, we show that the efficiency of repair is adversely affected by a deficiency in uPA or by the exogenous administration of PAI-1. By using PAI-1 variants and AEC from mice transgenically deficient in vitronectin (Vn), we demonstrate that the PAI-1 effect requires its Vn-binding activity. Furthermore, we have found that cell motility is enhanced by the availability of Vn in the matrix and that the AEC-Vn interaction is mediated, in part, by the alpha(v)beta(1) integrin. The significant effect of uPA and PAI-1 on epithelial repair suggests a mechanism by which the plasminogen system may modulate pulmonary fibrosis.


Assuntos
Bleomicina/análogos & derivados , Inibidor 1 de Ativador de Plasminogênio/farmacologia , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia , Vitronectina/metabolismo , Cicatrização/efeitos dos fármacos , Animais , Bleomicina/farmacologia , Linhagem Celular Tumoral , Movimento Celular , Colágeno/metabolismo , Humanos , Camundongos , Camundongos Knockout , Ligação Proteica , Alvéolos Pulmonares/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/farmacologia , Vitronectina/deficiência , Vitronectina/genética
12.
Am J Respir Cell Mol Biol ; 31(5): 552-8, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15284078

RESUMO

During inflammatory lung injury, the fibrinolytic activity that is normally present within bronchoalveolar lavage (BAL) fluid (BALF) is often suppressed due to increased levels of inhibitors, including plasminogen activator inhibitor (PAI)-1. Despite this suppression, BALF frequently contains fibrin degradation products, indicating persistence of fibrinolytic activity within the lung. To address this discrepancy and determine the sites where plasminogen activation is occurring, we developed an in situ zymographic technique for frozen sections of lung tissue that localizes plasminogen activator activity at the cellular level. After validating the method using enzyme inhibitors and mice with genetic manipulations of their plasminogen system genes, we applied the technique to lungs of normal and bleomycin-exposed mice. In normal mice, plasminogen activator activity was localized to bronchial epithelial cells, cells of the alveolar walls, and alveolar macrophages. After bleomycin exposure, in situ zymography showed that, despite loss of fibrinolytic activity within BALF, abundant enzymatic activity was associated with aggregates of inflammatory cells. PAI-1-deficient mice that are protected from bleomycin-induced fibrosis had preserved plasminogen activator activity in BALF and increased tissue activity, as determined by in situ zymography. We conclude that analysis of BALF does not adequately reflect the fibrinolytic activity that persists within microenvironments of the lung during inflammation.


Assuntos
Líquido da Lavagem Broncoalveolar , Pulmão/metabolismo , Ativadores de Plasminogênio/biossíntese , Animais , Antimetabólitos Antineoplásicos/farmacologia , Bleomicina/farmacologia , Relação Dose-Resposta a Droga , Células Epiteliais/metabolismo , Fibrinogênio/metabolismo , Fibrose/metabolismo , Humanos , Inflamação , Pulmão/citologia , Pneumopatias/patologia , Macrófagos Alveolares/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Plasminogênio/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ativadores de Plasminogênio/química , Ativadores de Plasminogênio/metabolismo , Fatores de Tempo
13.
Am J Pathol ; 163(2): 445-52, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12875966

RESUMO

Mice with homozygous deletion of the plasminogen activator inhibitor-1 gene (PAI-1(-/-)) are relatively protected from bleomycin-induced pulmonary fibrosis. At least part of the protective effect appears to occur during the latter stages of the pathological process when fibrotic tissue is being deposited. To investigate the effect of PAI-1 deficiency on fibrosis, we studied the accumulation of fibrotic tissue within subcutaneously implanted polyvinyl alcohol sponges. Similar to the effect of PAI-1 deficiency on bleomycin-induced pulmonary fibrosis, the accumulation of fibrotic tissue within implanted sponges occurred more slowly in PAI-1(-/-) compared to wild-type mice. Another striking difference observed in the PAI-1(-/-) mice was the rapid removal of a fibrin-rich matrix that formed within the sponges by 1 day after implantation in both wild-type and PAI-1(-/-) mice. The pattern of connective tissue invasion also differed: cells in wild-type mice infiltrated as individually penetrating cells whereas in PAI-1(-/-) mice they did so as a well-demarcated advancing front. Providing an alternative provisional matrix by impregnating sponges with a low concentration of collagen before implantation corrected the changes induced by PAI-1 deficiency. In conclusion, PAI-1 deficiency appears to affect fibrotic tissue formation in part by altering the provisional matrix that forms soon after tissue injury.


Assuntos
Fibrose/patologia , Pulmão/patologia , Inibidor 1 de Ativador de Plasminogênio/genética , Animais , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Fibrina/metabolismo , Fibrose/metabolismo , Corpos Estranhos , Hidroxiprolina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Álcool de Polivinil , Próteses e Implantes
14.
Am J Physiol Lung Cell Mol Physiol ; 283(5): L1023-32, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12376355

RESUMO

Plasminogen activator inhibitor-1 (PAI-1)-deficient transgenic mice have improved survival and less fibrosis after intratracheal bleomycin instillation. We hypothesize that PAI-1 deficiency limits scarring through unopposed plasminogen activation. If this is indeed true, then we would expect increased urokinase-type plasminogen activator (uPA) expression to result in a similar reduction in scarring and improvement in mortality. To test our hypothesis, using the tetracycline gene regulatory system, we have generated a transgenic mouse model with the features of inducible, lung-specific uPA production. After doxycycline administration, these transgenic animals expressed increased levels of uPA in their bronchoalveolar lavage (BAL) fluid that accelerated intrapulmonary fibrin clearance. Importantly, this increased plasminogen activator production led to a reduction in both lung collagen accumulation and mortality after bleomycin-induced injury. These results suggest that PAI-1 deficiency does protect against the effects of bleomycin-induced lung injury through unopposed plasmin generation. By allowing the manipulation of plasminogen activation at different phases of the fibrotic process, this model will serve as a powerful tool in further investigations into the pathogenesis of pulmonary fibrosis.


Assuntos
Regulação Enzimológica da Expressão Gênica/fisiologia , Lesão Pulmonar , Fibrose Pulmonar/prevenção & controle , Ativador de Plasminogênio Tipo Uroquinase/genética , Animais , Líquido da Lavagem Broncoalveolar/citologia , Primers do DNA , Genótipo , Humanos , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Transgênicos , Reação em Cadeia da Polimerase , Fibrose Pulmonar/genética , Fibrose Pulmonar/mortalidade , Fibrose Pulmonar/patologia , Taxa de Sobrevida , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA