Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Physiol Rep ; 7(4): e13977, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30806029

RESUMO

Endoglin (ENG) regulates signaling by transforming growth factor-ß (TGF-ß), a genetic modifier of cystic fibrosis (CF) lung disease severity. We hypothesized that ENG mediates TGF-ß pathobiology in CF airway epithelia. Comparing CF and non-CF human lungs, we measured ENG by qPCR, immunoblotting and ELISA. In human bronchial epithelial cell lines (16HBE), we used CFTR siRNA knockdown and functional inhibition (CFTRINH -172) to connect loss of CFTR to ENG synthesis. Plasmid overexpression of ENG assessed the direct effect of ENG on TGF-ß transcription and signal amplification in 16HBE cells. We found ENG protein to be increased more than fivefold both in human CF bronchoalveolar fluid (BALF) and human CF lung homogenates. ENG transcripts were increased threefold in CF, with a twofold increase in TGF-ß signaling. CFTR knockdown in 16HBE cells tripled ENG transcription and doubled protein levels with corresponding increases in TGF-ß signaling. Plasmid overexpression of ENG alone nearly doubled TGF-ß1 mRNA and increased TGF-ß signaling in 16HBE cells. These experiments identify that loss of CFTR function increases ENG expression in CF epithelia and amplifies TGF-ß signaling. Targeting ENG may offer a novel therapeutic opportunity to address TGF-ß associated pathobiology in CF.


Assuntos
Células Epiteliais Alveolares/metabolismo , Fibrose Cística/metabolismo , Endoglina/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Linhagem Celular , Células Cultivadas , Criança , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Endoglina/genética , Humanos , Transdução de Sinais
2.
Am J Respir Cell Mol Biol ; 56(3): 383-392, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27870560

RESUMO

Epidemiologic studies have linked gestational vitamin D deficiency to respiratory diseases, although mechanisms have not been defined. We hypothesized that antenatal vitamin D deficiency would impair airway development and alveolarization in a mouse model. We studied the effect of antenatal vitamin D deficiency by inducing it in pregnant mice and then compared lung development and function in their offspring to littermate controls. Postnatal vitamin D deficiency and sufficiency models from each group were also studied. We developed a novel tracheal ultrasound imaging technique to measure tracheal diameter in vivo. Histological analysis estimated tracheal cartilage total area and thickness. We found that vitamin D-deficient pups had reduced tracheal diameter with decreased tracheal cartilage minimal width. Vitamin D deficiency increased airway resistance and reduced lung compliance, and led to alveolar simplification. Postnatal vitamin D supplementation improved lung function and radial alveolar count, a parameter of alveolar development, but did not correct tracheal narrowing. We conclude that antenatal vitamin D deficiency impairs airway and alveolar development and limits lung function. Reduced tracheal diameter, cartilage irregularity, and alveolar simplification in vitamin D-deficient mice may contribute to increased airways resistance and diminished lung compliance. Vitamin D supplementation after birth improved lung function and, potentially, alveolar simplification, but did not improve defective tracheal structure. This mouse model offers insight into the mechanisms of vitamin D deficiency-associated lung disease and provides an in vivo model for investigating preclinical preventive and therapeutic strategies.


Assuntos
Traqueia/patologia , Deficiência de Vitamina D/patologia , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/patologia , Animais , Animais Recém-Nascidos , Feminino , Camundongos Endogâmicos C57BL , Gravidez , Testes de Função Respiratória , Traqueia/diagnóstico por imagem , Traqueia/efeitos dos fármacos , Traqueia/fisiopatologia , Vitamina D/farmacologia , Deficiência de Vitamina D/fisiopatologia
3.
Exp Gerontol ; 61: 62-75, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25451236

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a fatal lung disorder with unknown cause and no effective treatment. The incidence of and mortality from IPF increase with age, suggesting that advanced age is a major risk factor for IPF. The mechanism underlying the increased susceptibility of the elderly to IPF, however, is unknown. In this study, we show for the first time that the protein level of plasminogen activator inhibitor 1 (PAI-1), a protease inhibitor which plays an essential role in the control of fibrinolysis, was significantly increased with age in mouse lung homogenate and lung fibroblasts. Upon bleomycin challenge, old mice experienced augmented PAI-1 induction and lung fibrosis as compared to young mice. Most interestingly, we show that fewer (myo)fibroblasts underwent apoptosis and more (myo)fibroblasts with increased level of PAI-1 accumulated in the lung of old than in young mice after bleomycin challenge. In vitro studies further demonstrate that fibroblasts isolated from lungs of old mice were resistant to H2O2 and tumor necrosis factor alpha-induced apoptosis and had augmented fibrotic responses to TGF-ß1, compared to fibroblasts isolated from young mice. Inhibition of PAI-1 activity with a PAI-1 inhibitor, on the other hand, eliminated the aging-related apoptosis resistance and TGF-ß1 sensitivity in isolated fibroblasts. Moreover, we show that knocking down PAI-1 in human lung fibroblasts with PAI-1 siRNA significantly increased their sensitivity to apoptosis and inhibited their responses to TGF-ß1. Together, the results suggest that increased PAI-1 expression may underlie the aging-related sensitivity to lung fibrosis in part by protecting fibroblasts from apoptosis.


Assuntos
Envelhecimento , Apoptose , Fibroblastos/fisiologia , Inibidor 1 de Ativador de Plasminogênio/fisiologia , Fibrose Pulmonar/etiologia , Animais , Bleomicina/farmacologia , Líquido da Lavagem Broncoalveolar/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Crescimento Transformador beta1/fisiologia
4.
PLoS One ; 8(8): e70196, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23950911

RESUMO

RATIONALE: TGF-ß, a mediator of pulmonary fibrosis, is a genetic modifier of CF respiratory deterioration. The mechanistic relationship between TGF-ß signaling and CF lung disease has not been determined. OBJECTIVE: To investigate myofibroblast differentiation in CF lung tissue as a novel pathway by which TGF-ß signaling may contribute to pulmonary decline, airway remodeling and tissue fibrosis. METHODS: Lung samples from CF and non-CF subjects were analyzed morphometrically for total TGF-ß1, TGF-ß signaling (Smad2 phosphorylation), myofibroblast differentiation (α-smooth muscle actin), and collagen deposition (Masson trichrome stain). RESULTS: TGF-ß signaling and fibrosis are markedly increased in CF (p<0.01), and the presence of myofibroblasts is four-fold higher in CF vs. normal lung tissue (p<0.005). In lung tissue with prominent TGF-ß signaling, both myofibroblast differentiation and tissue fibrosis are significantly augmented (p<0.005). CONCLUSIONS: These studies establish for the first time that a pathogenic mechanism described previously in pulmonary fibrosis is also prominent in cystic fibrosis lung disease. The presence of TGF-ß dependent signaling in areas of prominent myofibroblast proliferation and fibrosis in CF suggests that strategies under development for other pro-fibrotic lung conditions may also be evaluated for use in CF.


Assuntos
Diferenciação Celular , Fibrose Cística/metabolismo , Pulmão/metabolismo , Miofibroblastos/citologia , Miofibroblastos/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Adulto , Fibrose Cística/patologia , Feminino , Fibrose , Humanos , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Pulmão/patologia , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Fosforilação , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
5.
PLoS One ; 5(7): e11662, 2010 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-20657842

RESUMO

Heterogeneous surface expression of Thy-1 in fibroblasts modulates inflammation and may thereby modulate injury and repair. As a paradigm, patients with idiopathic pulmonary fibrosis, a disease with pathologic features of chronic inflammation, demonstrate an absence of Thy-1 immunoreactivity within areas of fibrotic activity (fibroblast foci) in contrast to the predominant Thy-1 expressing fibroblasts in the normal lung. Likewise, Thy-1 deficient mice display more severe lung fibrosis in response to an inflammatory injury than wildtype littermates. We investigated the role of Thy-1 in the response of fibroblasts to the pro-inflammatory cytokine TNF-alpha. Our study demonstrates distinct profiles of TNF-alpha-activated gene expression in Thy-1 positive (Thy-1+) and negative (Thy-1-) subsets of mouse embryonic fibroblasts (MEF). TNF-alpha induced a robust activation of MMP-9, ICAM-1, and the IL-8 promoter driven reporter in Thy-1- MEFs, in contrast to only a modest increase in Thy-1+ counterparts. Consistently, ectopic expression of Thy-1 in Thy-1- MEFs significantly attenuated TNF-alpha-activated gene expression. Mechanistically, TNF-alpha activated Src family kinase (SFK) only in Thy-1- MEFs. Blockade of SFK activation abrogated TNF-alpha-activated gene expression in Thy-1- MEFs, whereas restoration of SFK activation rescued the TNF-alpha response in Thy-1+ MEFs. Our findings suggest that Thy-1 down-regulates TNF-alpha-activated gene expression via interfering with SFK- and NF-kappaB-mediated transactivation. The current study provides a novel mechanistic insight to the distinct roles of fibroblast Thy-1 subsets in inflammation.


Assuntos
Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Antígenos Thy-1/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Quinases da Família src/metabolismo , Animais , Células Cultivadas , Citometria de Fluxo , Immunoblotting , Molécula 1 de Adesão Intercelular/genética , Interleucina-8/genética , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Mutantes , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Antígenos Thy-1/genética , Quinases da Família src/genética
6.
Am J Physiol Lung Cell Mol Physiol ; 296(5): L738-50, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19270178

RESUMO

Transforming growth factor (TGF)-beta mediates hypoxia-induced inhibition of alveolar development in the newborn lung. TGF-beta is regulated primarily at the level of activation of latent TGF-beta. Fibroblasts expressing Thy-1 (CD90) inhibit TGF-beta activation. We hypothesized that loss of Thy-1 due to hypoxia may be a mechanism by which hypoxia increases TGF-beta activation and that animals deficient in Thy-1 will simulate the effects of hypoxia on lung development. To determine if loss of Thy-1 occurred during hypoxia, non-transgenic (C57BL/6) wild-type (WT) mice exposed to hypoxia were evaluated for Thy-1 mRNA and protein. To determine if Thy-1 deficiency simulated hypoxia, WT and Thy-1 null (Thy-1(-/-)) mice were exposed to air or hypoxia from birth to 2 wk, the critical period of lung development, and lung histology, function, parameters related to TGF-beta signaling, and extracellular matrix protein content were measured. To test if the phenotype in Thy-1(-/-) mice was due to excessive TGF-beta signaling, measurements were also performed in Thy-1(-/-) mice administered TGF-beta neutralizing antibody (1D11). We observed that hypoxia reduced Thy-1 mRNA and Thy-1 staining in WT mice. Thy-1(-/-) mice had impaired alveolarization, increased TGF-beta signaling, reduced lung epithelial and endothelial cell proliferation but increased fibroblast proliferation, and increased collagen and elastin. Lung compliance was lower, and tissue but not airway resistance was higher in Thy-1(-/-) mice at 2 wk. Thy-1(-/-) mice given 1D11 had improved alveolar development and lung function. These data support the hypothesis that hypoxia, by reducing Thy-1, increases TGF-beta activation, and thereby inhibits normal alveolar development.


Assuntos
Alvéolos Pulmonares/crescimento & desenvolvimento , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo , Ar , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Hipóxia Celular , Proliferação de Células , Colágeno/metabolismo , Elastina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Alvéolos Pulmonares/patologia , Alvéolos Pulmonares/ultraestrutura , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Testes de Função Respiratória , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
7.
Am J Pathol ; 167(2): 365-79, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16049324

RESUMO

Fibroblasts consist of heterogeneous subpopulations that have distinct roles in fibrotic responses. Previously we reported enhanced proliferation in response to fibrogenic growth factors and selective activation of latent transforming growth factor (TGF)-beta in fibroblasts lacking cell surface expression of Thy-1 glycoprotein, suggesting that Thy-1 modulates the fibrogenic potential of fibroblasts. Here we report that compared to controls Thy-1-/- C57BL/6 mice displayed more severe histopathological lung fibrosis, greater accumulation of lung collagen, and increased TGF-beta activation in the lungs 14 days after intratracheal bleomycin. The majority of cells demonstrating TGF-beta activation and myofibroblast differentiation in bleomycin-induced lesions were Thy-1-negative. Histological sections from patients with idiopathic pulmonary fibrosis demonstrated absent Thy-1 staining within fibroblastic foci. Normal lung fibroblasts, in both mice and humans, were predominantly Thy-1-positive. The fibrogenic cytokines interleukin-1 and tumor necrosis factor-alpha induced loss of fibroblast Thy-1 surface expression in vitro, which was associated with Thy-1 shedding, Smad phosphorylation, and myofibroblast differentiation. These results suggest that fibrogenic injury promotes loss of lung fibroblast Thy-1 expression, resulting in enhanced fibrogenesis.


Assuntos
Fibroblastos/metabolismo , Pulmão/metabolismo , Fibrose Pulmonar/metabolismo , Antígenos Thy-1/fisiologia , Animais , Antimetabólitos Antineoplásicos/farmacologia , Bleomicina/farmacologia , Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Humanos , Interleucina-1/metabolismo , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso/efeitos dos fármacos , Músculo Liso/metabolismo , Fosforilação , Fibrose Pulmonar/patologia , Proteína Smad3 , Antígenos Thy-1/genética , Transativadores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
8.
Exp Cell Res ; 295(2): 488-96, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15093746

RESUMO

The precise biological role of Thy-1, a glycophosphatidyl-inositol (GPI)-linked cell surface glycoprotein in non-caveolar lipid raft microdomains, remains enigmatic. Evidence suggests that Thy-1 affects intracellular signaling through src-family protein kinases, and modulates adhesive and migratory events, such as thymocyte adhesion and neurite extension. Primary fibroblasts sorted based on presence or absence of cell surface Thy-1 display strikingly distinct morphologies and differ with respect to production of and response to cytokines and growth factors. It is unclear the extent to which Thy-1 mediates these differences. Findings reported here indicate a novel role for Thy-1 in regulating the activity of Rho GTPase, a critical regulator of cellular adhesion and cytoskeletal organization. Endogenous or heterologous Thy-1 expression promotes focal adhesion and stress fiber formation, characteristic of increased Rho GTPase activity, and inhibits migration. Immunoblotting following transfection of RFL6 fibroblasts with Thy-1 demonstrates that Thy-1 expression inhibits src-family protein tyrosine kinase (SFK) activation, resulting in decreased phosphorylation of p190 Rho GTPase-activating protein (GAP). This results in a net increase in active Rho, and increased stress fibers and focal adhesions. We therefore conclude that Thy-1 surface expression regulates fibroblast focal adhesions, cytoskeletal organization and migration by modulating the activity of p190 RhoGAP and Rho GTPase.


Assuntos
Movimento Celular , Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Adesões Focais/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Nucleares/metabolismo , Antígenos Thy-1/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Células Cultivadas , Proteínas de Ligação a DNA , Ativação Enzimática , Fibroblastos/enzimologia , Proteínas Ativadoras de GTPase , Pulmão/citologia , Camundongos , Camundongos Knockout , Modelos Biológicos , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Ratos , Ratos Endogâmicos Lew , Proteínas Repressoras , Quinases da Família src/antagonistas & inibidores
9.
J Biol Chem ; 279(22): 23510-6, 2004 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-15033989

RESUMO

The hep I peptide of thrombospondin-1 is known to induce the disassembly of focal adhesions, a critical step in regulating cellular adhesive changes needed for cell motility. Fibroblasts that are heterogeneous with respect to the surface expression of Thy-1 differ markedly in morphology, cytoskeletal organization, and migration, suggesting differential regulation of focal adhesion dynamics. Here we demonstrate that disassembly of focal adhesions mediated by both full-length thrombospondin-1 and the hep I peptide in fibroblasts requires the expression of Thy-1, although it does not appear to function as a stable member of the hep I receptor complex. Consistent with a known function of Thy-1 in regulating lipid raft-associated signaling, intact lipid rafts are necessary for hep I-mediated focal adhesion disassembly. Furthermore, we establish Src family kinase (SFK) activation as a novel component required for hep I-induced signaling leading to focal adhesion disassembly. hep I induces transient phosphorylation of SFKs in Thy-1-expressing fibroblasts only. Therefore, we conclude that Thy-1 surface expression is required for thrombospondin-1-induced focal adhesion disassembly in fibroblasts through an SFK-dependent mechanism. This represents a novel role for Thy-1 in the regulation of fibroblast-matrix interactions critical to tissue homeostasis and remodeling.


Assuntos
Adesões Focais/fisiologia , Trombospondina 1/fisiologia , Antígenos Thy-1/biossíntese , Animais , Ativação Enzimática , Fibroblastos/citologia , Fibroblastos/fisiologia , Microdomínios da Membrana/fisiologia , Ratos , Transdução de Sinais , Quinases da Família src/metabolismo
10.
Am J Respir Cell Mol Biol ; 26(6): 702-8, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12034569

RESUMO

Following lung injury or inflammation, fibroblasts mediate either restorative repair or disordered remodeling. Interleukin (IL)-1beta is a key mediator in the transition from injury/inflammation to tissue remodeling, in part through its regulation of platelet-derived growth factor alpha receptor (PDGFalphaR). Based on prior demonstration of differential PDGFalphaR expression, we hypothesized that subpopulations of fibroblasts would have heterogeneous responses to IL-1. We report that IL-1beta significantly increases expression of PDGFalphaR in Thy-1-, but not Thy-1+ fibroblasts. Higher baseline expression of PDGFalphaR in Thy-1- fibroblasts is partially abrogated by IL-1 receptor antagonist. There are no differences in IL-1beta binding, as determined by flow cytometry, or in the presence of the type I IL-1 receptor (IL-1RtI) or its associated protein (IL-1RacP) by immunoblotting. IL-1beta induces DNA binding of both nuclear factor kappaB (NF-kappaB) and CAATT-enhancer binding protein (C/EBP), and activation of p38 mitogen-activated protein kinase in both subpopulations. However, IL-1beta-induced proliferation and expression of IL-6 are significantly higher in Thy-1- fibroblasts. Heterogeneous responses to IL-1beta despite equivalent presence of both proximal and distal signaling components indicates that parallel signaling pathways are activated selectively in Thy-1- cells, suggesting a prominent role for this subset in the transition from inflammation to lung remodeling.


Assuntos
Interleucina-1/fisiologia , Pulmão/metabolismo , Transdução de Sinais/fisiologia , Animais , Sequência de Bases , Western Blotting , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Divisão Celular/fisiologia , DNA/metabolismo , Primers do DNA , Fibroblastos/metabolismo , Citometria de Fluxo , Pulmão/citologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Fosforilação , Ratos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de Interleucina-1/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...