Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Respir Res ; 14: 80, 2013 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-23915349

RESUMO

BACKGROUND: Interstitial lung disease is a major cause of morbidity and mortality in systemic sclerosis (SSc), with insufficiently effective treatment options. Progression of pulmonary fibrosis involves expanding populations of fibroblasts, and the accumulation of extracellular matrix proteins. Characterisation of SSc lung fibroblast gene expression profiles underlying the fibrotic cell phenotype could enable a better understanding of the processes leading to the progressive build-up of scar tissue in the lungs. In this study we evaluate the transcriptomes of fibroblasts isolated from SSc lung biopsies at the time of diagnosis, compared with those from control lungs. METHODS: We used Affymetrix oligonucleotide microarrays to compare the gene expression profile of pulmonary fibroblasts cultured from 8 patients with pulmonary fibrosis associated with SSc (SSc-ILD), with those from control lung tissue peripheral to resected cancer (n=10). Fibroblast cultures from 3 patients with idiopathic pulmonary fibrosis (IPF) were included as a further comparison. Genes differentially expressed were identified using two separate analysis programs following a set of pre-determined criteria: only genes significant in both analyses were considered. Microarray expression data was verified by qRT-PCR and/or western blot analysis. RESULTS: A total of 843 genes were identified as differentially expressed in pulmonary fibroblasts from SSc-ILD and/or IPF compared to control lung, with a large overlap in the expression profiles of both diseases. We observed increased expression of a TGF-ß response signature including fibrosis associated genes and myofibroblast markers, with marked heterogeneity across samples. Strongly suppressed expression of interferon stimulated genes, including antiviral, chemokine, and MHC class 1 genes, was uniformly observed in fibrotic fibroblasts. This expression profile includes key regulators and mediators of the interferon response, such as STAT1, and CXCL10, and was also independent of disease group. CONCLUSIONS: This study identified a strongly suppressed interferon-stimulated gene program in fibroblasts from fibrotic lung. The data suggests that the repressed expression of interferon-stimulated genes may underpin critical aspects of the profibrotic fibroblast phenotype, identifying an area in pulmonary fibrosis that requires further investigation.


Assuntos
Fibroblastos/metabolismo , Interferons/metabolismo , Doenças Pulmonares Intersticiais/complicações , Doenças Pulmonares Intersticiais/metabolismo , Pulmão/metabolismo , Escleroderma Sistêmico/complicações , Escleroderma Sistêmico/metabolismo , Adulto , Idoso , Células Cultivadas , Regulação para Baixo , Feminino , Perfilação da Expressão Gênica , Humanos , Pulmão/patologia , Doenças Pulmonares Intersticiais/patologia , Masculino , Pessoa de Meia-Idade , Análise Serial de Proteínas , Escleroderma Sistêmico/patologia
2.
Arthritis Rheum ; 62(5): 1523-32, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20213804

RESUMO

OBJECTIVE: Connective tissue growth factor (CTGF) is a cysteine-rich secreted matricellular protein involved in wound healing and tissue repair. Enhanced and prolonged expression of CTGF has been associated with tissue fibrosis in humans. However, questions remain as to whether CTGF expression alone is sufficient to drive fibrosis. This study was undertaken to investigate whether CTGF alone is sufficient to cause fibrosis in intact animals and whether its effects are mediated through activation of transforming growth factor beta (TGFbeta) signaling or through distinct signal transduction pathways. METHODS: We generated mice overexpressing CTGF in fibroblasts under the control of the fibroblast-specific collagen alpha2(I) promoter enhancer. Tissues such as skin, lung, and kidney were harvested for histologic analysis. Mouse embryonic fibroblasts were prepared from embryos (14.5 days postcoitum) for biochemical analysis. RESULTS: Mice overexpressing CTGF in fibroblasts were susceptible to accelerated tissue fibrosis affecting the skin, lung, kidney, and vasculature, most notably the small arteries. We identified a marked expansion of the myofibroblast cell population in the dermis. RNA analysis of transgenic dermal fibroblasts revealed elevated expression of key matrix genes, consistent with a fibrogenic response. CTGF induced phosphorylation of p38, ERK-1/2, JNK, and Akt, but not Smad3, in transgenic mouse fibroblasts compared with wild-type mouse fibroblasts. Transfection experiments showed significantly increased basal activity of the CTGF and serum response element promoters, and enhanced induction of the CTGF promoter in the presence of TGFbeta. CONCLUSION: These results demonstrate that selective expression of CTGF in fibroblasts alone causes tissue fibrosis in vivo through specific signaling pathways, integrating cues from the extracellular matrix into signal transduction pathways to orchestrate pivotal biologic responses relevant to tissue repair and fibrosis.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fibroblastos/patologia , Cirrose Hepática , Fibrose Pulmonar , Animais , Membrana Basal/patologia , Vasos Sanguíneos/patologia , Diferenciação Celular/fisiologia , Colágeno/genética , Colágeno Tipo I , Fibroblastos/metabolismo , Expressão Gênica/fisiologia , Glomerulosclerose Segmentar e Focal/metabolismo , Glomerulosclerose Segmentar e Focal/patologia , Glomerulosclerose Segmentar e Focal/fisiopatologia , Rim/patologia , Óperon Lac , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Cirrose Hepática/fisiopatologia , Linfocinas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Regiões Promotoras Genéticas/fisiologia , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Fibrose Pulmonar/fisiopatologia , Transdução de Sinais/fisiologia , Pele/patologia , Fator de Crescimento Transformador beta/metabolismo
3.
N Engl J Med ; 357(12): 1210-20, 2007 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-17881752

RESUMO

BACKGROUND: Systemic sclerosis (scleroderma) is a life-threatening autoimmune disease that is characterized by the presence of specific autoantibodies and fibrosis of the skin and major internal organs. METHODS: We genotyped a polymorphism (G-945C) in the promoter of the connective-tissue growth factor (CTGF) gene in 1000 subjects in two groups: group 1, consisting of 200 patients with systemic sclerosis and 188 control subjects; and group 2, consisting of 300 patients with systemic sclerosis and 312 control subjects. The combined groups represented an estimated 10% of patients with systemic sclerosis in the United Kingdom. We tested the effect of the polymorphism on the transcription of CTGF. RESULTS: The GG genotype was significantly more common in patients with systemic sclerosis than in control subjects in both groups, with an odds ratio for the combined group of 2.2 (95% confidence interval [CI], 1.5 to 3.2; P<0.001 for trend). Analysis of the combined group of patients with systemic sclerosis showed a significant association between homozygosity for the G allele and the presence of anti-topoisomerase I antibodies (odds ratio, 3.3; 95% CI, 2.0 to 5.6; P<0.001) and fibrosing alveolitis (odds ratio, 3.1; 95% CI, 1.9 to 5.0; P<0.001). We observed that the substitution of cytosine for guanine created a binding site of the transcriptional regulators Sp1 and Sp3. The C allele has high affinity for Sp3 and is associated with severely reduced transcriptional activity. A chromatin immunoprecipitation assay showed a marked shift in the ratio of Sp1 to Sp3 binding at this region, demonstrating functional relevance in vivo. CONCLUSIONS: The G-945C substitution represses CTGF transcription, and the -945G allele is significantly associated with susceptibility to systemic sclerosis.


Assuntos
Predisposição Genética para Doença , Proteínas Imediatamente Precoces/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Mutação Puntual , Regiões Promotoras Genéticas , Escleroderma Sistêmico/genética , Estudos de Casos e Controles , Fator de Crescimento do Tecido Conjuntivo , Feminino , Genótipo , Humanos , Masculino , Reação em Cadeia da Polimerase , Polimorfismo Genético , Polimorfismo de Nucleotídeo Único , Fatores de Risco , Análise de Sequência de DNA , Transcrição Gênica
4.
J Vasc Surg ; 51(3): 689-99, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20206813

RESUMO

PURPOSE: Critical leg ischemia (CLI) is associated with a high morbidity and mortality. Therapeutic angiogenesis is still being investigated as a possible alternative treatment option for CLI. CXCL12, a chemokine, is known to have two spliced variants, CXCL12alpha and CXCL12beta, but the significance remains unknown. The study investigated the angiogenic effects of CXCL12, protein expressions of CXCL12, and the receptor CXCR4 in human CLI. METHODS: In vitro, human microvascular endothelial cells (HMEC-1) were used. Cell proliferation was assessed using methylene blue assay and cell count method. Apoptosis was determined by counting the pyknotic nuclei after 4'-6-diamidino-2-phenylindole staining and confirmed by caspase-3 assay. We employed matrigel as capillary tube formation assay. The activity of signaling pathways was measured using Western blotting. In vivo, gastrocnemius biopsies were obtained from the lower limbs of patients with CLI and controls (n = 12 each). Immunohistochemistry, double immunofluorescence labeling, and Western blotting were then performed. RESULTS: CXCL12 attenuated HMEC-1 apoptosis (P < .01), stimulated cell proliferation (P < .05) and capillary tube formation (P < .01). Compared with CXCL12alpha, CXCL12beta has a greater effect on apoptosis and cell proliferation (P < .01). Treatment with both variants resulted in time-dependent activation of PI3K/Akt and p44/42 but not p38 MAP kinase. In CLI, CXCL12alpha was expressed by skeletal muscle fibers with minimal expression of CXCL12beta. CXCR4 was extensively expressed and colocalized to microvessels. A significant 2.6-fold increase in CXCL12alpha and CXCR4 expressions (P < .01) were noted in CLI but not for CXCL12beta (P > .05). CONCLUSIONS: The study showed that CXCL12beta had more potent angiogenic properties but was not elevated in human CLI biopsies. This provided an interesting finding on the role of CXCL12 variants in pathophysiologic angiogenic response in CLI.


Assuntos
Quimiocina CXCL12/metabolismo , Células Endoteliais/imunologia , Isquemia/imunologia , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Receptores CXCR4/metabolismo , Idoso , Indutores da Angiogênese/farmacologia , Apoptose , Biópsia , Western Blotting , Estudos de Casos e Controles , Proliferação de Células , Células Cultivadas , Quimiocina CXCL12/farmacologia , Estado Terminal , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Feminino , Imunofluorescência , Humanos , Isquemia/patologia , Isquemia/fisiopatologia , Extremidade Inferior , Masculino , Pessoa de Meia-Idade , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Músculo Esquelético/imunologia , Neovascularização Fisiológica/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Isoformas de Proteínas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Fatores de Tempo
5.
Biomed Res Int ; 2019: 1484736, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31119153

RESUMO

BACKGROUND AND OBJECTIVE: Progressive pulmonary fibrosis is the main cause of death in patients with systemic sclerosis (SSc) with interstitial lung disease (ILD) and in those with idiopathic pulmonary fibrosis (IPF). Transforming growth factor-ß (TGF-ß) and NADPH oxidase- (NOX-) derived reactive oxygen species (ROS) are drivers of lung fibrosis. We aimed to determine the role of the epigenetic readers, bromodomain and extraterminal (BET) proteins in the regulation of redox balance in activated myofibroblasts. METHODS: In TGF-ß-stimulated fibroblasts, we investigated the effect of the BET inhibitor JQ1 on the mRNA expression of the prooxidant gene NOX4 and the antioxidant gene superoxide dismutase (SOD2) by quantitative RT-PCR, the antioxidant transcription factor NF-E2-related factor 2 (Nrf2) activity by a reporter assay, and intracellular ROS levels by dichlorofluorescein staining. Myofibroblast activation was determined by α-smooth muscle actin immunocytochemistry. The role of specific BET protein isoforms in NOX4 gene regulation was studied by siRNA silencing and chromatin-immunoprecipitation. RESULTS AND CONCLUSIONS: Affymetrix gene array analysis revealed increased NOX4 and reduced SOD2 expression in SSc and IPF fibroblasts. SOD2 silencing in non-ILD control fibroblasts induced a profibrotic phenotype. TGF-ß increased NOX4 and inhibited SOD2 expression, while increasing ROS production and myofibroblast differentiation. JQ1 reversed the TGF-ß-mediated NOX4/SOD2 imbalance and Nrf2 inactivation and attenuated ROS production and myofibroblast differentiation. The BET proteins Brd3 and Brd4 were shown to bind to the NOX4 promoter and drive TGF-ß-induced NOX4 expression. Our data indicate a critical role of BET proteins in promoting redox imbalance and pulmonary myofibroblast activation and support BET bromodomain inhibitors as a potential therapy for fibrotic lung disease.


Assuntos
Fibrose Pulmonar Idiopática/genética , NADPH Oxidase 4/genética , Proteínas Nucleares/genética , Proteínas de Ligação a RNA/genética , Escleroderma Sistêmico/genética , Fatores de Transcrição/genética , Azepinas/farmacologia , Proteínas de Ciclo Celular , Desdiferenciação Celular/genética , Proliferação de Células/genética , Epigênese Genética , Regulação da Expressão Gênica/genética , Humanos , Fibrose Pulmonar Idiopática/complicações , Fibrose Pulmonar Idiopática/patologia , Pulmão/metabolismo , Pulmão/patologia , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Fator 2 Relacionado a NF-E2/genética , Oxirredução , RNA Interferente Pequeno/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Escleroderma Sistêmico/complicações , Escleroderma Sistêmico/patologia , Superóxido Dismutase/genética , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/farmacologia , Triazóis/farmacologia
6.
J Invest Dermatol ; 129(1): 194-204, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18563179

RESUMO

Previous attempts to delete type II TGFbeta receptor (TbetaRII) in fibroblasts have precluded examination of adult mice due to early mortality. We have selectively deleted TbetaRII postnatally in differentiated connective tissue fibroblasts using an inducible Cre-Lox strategy. Tamoxifen-dependent Cre recombinase linked to a fibroblast-specific regulatory sequence from the proalpha2(I)collagen gene permitted deletion of floxed TbetaRII alleles. After postnatal deletion of TbetaRII in fibroblasts, healing of excisional skin wounds in adults showed markedly attenuated dermal scar formation, defective wound contraction and enhanced epidermal proliferation. These findings support a pivotal role for transforming growth factor beta (TGFbeta) signalling in fibroblasts in regulating normal skin wound healing. Explanted dermal fibroblasts from TbetaRII-null-fib mice showed impaired migration and did not generate normal contractile biomechanical forces in fixed collagen gels nor develop alpha-smooth muscle antigen-rich stress fibers in response to TGFbeta1. Surprisingly, some TGFbeta-regulated proteins, including connective tissue growth factor (CTGF), were basally upregulated in TbetaRII-null fibroblasts and this was dependent on extracellular signal-regulated kinase 1/2 activity in these cells. This suggests that other intracellular pathways regulating CTGF expression may partially compensate for disruption of TGFbeta signalling in fibroblasts. Together, our data confirm that expression of TbetaRII in differentiated dermal fibroblasts is essential for normal wound healing and demonstrate a critical role in the development and function of myofibroblasts.


Assuntos
Fibroblastos/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Pele/patologia , Alelos , Animais , Fenômenos Biomecânicos , Linhagem da Célula , Citoesqueleto , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Deleção de Genes , Camundongos , Músculos/citologia , Receptor do Fator de Crescimento Transformador beta Tipo II , Transdução de Sinais , Cicatrização
7.
Arthritis Rheum ; 60(7): 2142-55, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19565505

RESUMO

OBJECTIVE: Connective tissue growth factor (CTGF; CCN2) is overexpressed in systemic sclerosis (SSc) and has been hypothesized to be a key mediator of the pulmonary fibrosis frequently observed in this disease. CTGF is induced by transforming growth factor beta (TGFbeta) and is a mediator of some profibrotic effects of TGFbeta in vitro. This study was undertaken to investigate the role of CTGF in enhanced expression of type I collagen in bleomycin-induced lung fibrosis, and to delineate the mechanisms of action underlying the effects of CTGF on Col1a2 (collagen gene type I alpha2) in this mouse model and in human pulmonary fibroblasts. METHODS: Transgenic mice that were carrying luciferase and beta-galactosidase reporter genes driven by the Col1a2 enhancer/promoter and the CTGF promoter, respectively, were injected with bleomycin to induce lung fibrosis (or saline as control), and the extracted pulmonary fibroblasts were incubated with CTGF blocking agents. In vitro, transient transfection, promoter/reporter constructs, and electrophoretic mobility shift assays were used to determine the mechanisms of action of CTGF in pulmonary fibroblasts. RESULTS: In the mouse lung tissue, CTGF expression and promoter activity peaked 1 week after bleomycin challenge, whereas type I collagen expression and Col1a2 promoter activity peaked 2 weeks postchallenge. Fibroblasts isolated from the mouse lungs 14 days after bleomycin treatment retained a profibrotic expression pattern, characterized by greatly elevated levels of type I collagen and CTGF protein and increased promoter activity. In vitro, inhibition of CTGF by specific small interfering RNA and neutralizing antibodies reduced the collagen protein expression and Col1a2 promoter activity. Moreover, in vivo, anti-CTGF antibodies applied after bleomycin challenge significantly reduced the Col1a2 promoter activity by approximately 25%. The enhanced Col1a2 promoter activity in fibroblasts from bleomycin-treated lungs was partly dependent on Smad signaling, whereas CTGF acted on the Col1a2 promoter by a mechanism that was independent of the Smad binding site, but was, instead, dependent on the ERK-1/2 and JNK MAPK pathways. The CTGF effect was mapped to the proximal promoter region surrounding the inverted CCAAT box, possibly involving CREB and c-Jun. In human lung fibroblasts, the human COL1A2 promoter responded in a similar manner, and the mechanisms of action also involved ERK-1/2 and JNK signaling. CONCLUSION: Our results clearly define a direct profibrotic effect of CTGF and demonstrate its contribution to lung fibrosis through transcriptional activation of Col1a2. Blocking strategies revealed the signaling mechanisms involved. These findings show CTGF to be a rational target for therapy in fibrotic diseases such as SSc.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fibroblastos/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fibrose Pulmonar/metabolismo , Transcrição Gênica/fisiologia , Animais , Sequência de Bases , Bleomicina/efeitos adversos , Células Cultivadas , Colágeno/metabolismo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Fator de Crescimento do Tecido Conjuntivo/antagonistas & inibidores , Fator de Crescimento do Tecido Conjuntivo/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Modelos Animais de Doenças , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Humanos , MAP Quinase Quinase 4/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/fisiologia , Proteínas Smad/metabolismo , Transcrição Gênica/efeitos dos fármacos
8.
Growth Factors ; 26(2): 80-91, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18428027

RESUMO

Connective tissue growth factor (CTGF) is a member of the CCN family of six small secreted, cysteine-rich growth factors. The unique modular structure encompasses distinct functional domains which enable CTGF to interact with growth factors, surface receptors and matrix components. Widely expressed, CTGF has critical roles in embryonic development and the maintenance of normal cell and connective tissue function. It is also important for tissue repair following injury, and has been implicated in common diseases including atherosclerosis, pulmonary and renal fibrotic disorders and cancer. Factors that regulate CTGF transcription in response to exogenous stimuli, as well as downstream signalling pathways, have been described. However, only recently have studies begun to unravel how the functional domains within the CTGF modules orchestrate signals and control key biological processes. This article highlights how the structural and functional domains of CTGF and CTGF cleavage fragments integrate multiple extracellular events into cell signals.


Assuntos
Proteínas Imediatamente Precoces/química , Proteínas Imediatamente Precoces/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/química , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Animais , Fenômenos Fisiológicos Celulares , Células/citologia , Células/metabolismo , Fator de Crescimento do Tecido Conjuntivo , Humanos , Proteínas Imediatamente Precoces/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Neoplasias/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Relação Estrutura-Atividade
9.
Arthritis Rheum ; 58(4): 1175-88, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18383385

RESUMO

OBJECTIVE: To explore increased susceptibility to fibrosis following experimental injury to alveolar epithelial cells (AECs) in a novel transgenic mouse model of scleroderma with fibroblast-specific perturbation of transforming growth factor beta (TGFbeta) signaling (TbetaRIIDeltak-fib mice). METHODS: Wild-type (WT) and transgenic mice were injured with intratracheally administered saline or bleomycin, and the lungs were harvested for biochemical, histologic, and electron microscopic analysis. RESULTS: Electron microscopy revealed AEC abnormalities in the lungs of untreated transgenic mice and bleomycin-treated WT mice; the lungs of transgenic mice treated with bleomycin showed severe epithelial damage. Compared with lungs from bleomycin-treated WT mice, lungs from bleomycin-treated transgenic mice demonstrated increased fibroproliferation, myofibroblast persistence, and impaired hyperplasia and increased apoptosis of type II AECs. The lungs from saline-treated transgenic mice and those from bleomycin-treated WT mice had phenotypic similarities, suggesting enhanced susceptibility to minor epithelial injury in the transgenic strain. The level of collagen was increased in the lungs from transgenic mice compared with that in the lungs from WT mice after treatment with either bleomycin or saline. Persistent fibrosis in bleomycin-treated transgenic mice was independent of ongoing neutrophil inflammation but was associated with impaired alveolar epithelial repair. CONCLUSION: These results suggest that in the context of fibroblast-specific perturbation of TGFbeta signaling, even minor epithelial injury induces significant fibrosis. The model supports a central role for TGFbeta in determining fibrosis and demonstrates that lung fibroblasts may regulate the response of AECs to injury. Our findings provide insight into likely pathogenic mechanisms in scleroderma-associated pulmonary fibrosis.


Assuntos
Fibroblastos/metabolismo , Fibrose Pulmonar/complicações , Fibrose Pulmonar/fisiopatologia , Escleroderma Sistêmico , Fator de Crescimento Transformador beta/metabolismo , Animais , Bleomicina/administração & dosagem , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais , Irritantes/administração & dosagem , Camundongos , Camundongos Transgênicos , Alvéolos Pulmonares/citologia , Fibrose Pulmonar/induzido quimicamente , Escleroderma Sistêmico/complicações , Escleroderma Sistêmico/fisiopatologia , Transdução de Sinais , Cloreto de Sódio/administração & dosagem
10.
J Vasc Surg ; 43(1): 125-33, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16414399

RESUMO

OBJECTIVE: The potent physiologic endogenous angiogenic response to ischemic stimuli is often suboptimal, and therefore, a better understanding of the basic mechanisms is essential for the use in therapeutic angiogenesis. Hypoxia-inducible factor-1 (HIF-1) is a major transcription factor that promotes ischemia-driven angiogenesis and is induced when the HIF-1alpha subunit is upregulated. However, little is known about the endogenous angiogenic response and the role of HIF-1alpha in human critical limb ischemia (CLI). We aimed to investigate the extent of the angiogenic response and the expression of HIF-1alpha in the lower limbs of CLI patients. METHOD: Skeletal muscle biopsy specimens were obtained from the lower limbs of 12 patients with CLI and 12 patients without limb ischemia (controls), with ethical committee approval. Microvessel density (MVD) was determined by using endothelial marker anti-CD31, and HIF-1alpha expressions were determined by immunohistochemistry. MVD was measured as the median number of microvessels in x200 magnification fields. Five random fields per section and three sections per biopsy specimen were analyzed. Enzyme-linked immunoabsorbent assay and Western blotting were used to quantify the HIF-1alpha levels. Colocalization between cell-specific antigens was investigated by double immunofluorescence labelling by using confocal microscopy. Statistical analyses were performed with the Mann-Whitney U test. RESULTS: The CLI group have significantly higher MVD, with an increase of 2.7-fold compared with the controls (P < 0.001). HIF-1alpha expression was significantly increased in CLI muscles (P < 0.001) and was localized to vascular endothelial cells. CONCLUSIONS: Our findings suggest that the endogenous angiogenic response occurs in CLI. The increased HIF-1alpha level and colocalization to vascular endothelial cells suggest that HIF-1alpha plays a role in the physiologic endogenous angiogenic response in CLI. Therefore, augmentation of the HIF-1alpha pathway may be an important aspect in therapeutic angiogenesis.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Isquemia/metabolismo , Isquemia/patologia , Perna (Membro)/irrigação sanguínea , Neovascularização Patológica , Idoso , Estado Terminal , Feminino , Humanos , Masculino , Microcirculação
11.
Am J Pathol ; 169(6): 2254-65, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17148686

RESUMO

Connective tissue remodeling provides mammals with a rapid mechanism to repair wounds after injury. Inappropriate activation of this reparative process leads to scarring and fibrosis. Here, we studied the effects of platelet-derived growth factor receptor-beta blockade in vivo using the platelet-derived growth factor receptor (PDGFR)-beta inhibitor imatinib mesylate on tissue repair. After 7 days, healing of wounds was delayed with significantly reduced wound closure and concomitant reduction in myofibroblast frequency, expression of fibronectin ED-A, and collagen type I. Using a collagen type I transgenic reporter mouse, we showed that inhibiting PDGFR-beta activation restricted the distribution of collagen-synthesizing cells to wound margins and dramatically reduced cell proliferation in vivo. By 14 days, treated wounds were fully closed. Blocking PDGFR-beta signaling did not prevent the differentiation of myofibroblasts in vitro but potently inhibited fibroblast proliferation and migration. In addition, PDGFR-beta inhibition in vivo was accompanied by abnormal microvascular morphogenesis reminiscent of that observed in PDGFR-beta-/- mice with significantly reduced immunostaining of the pericyte marker NG2. Imatinib treatment also inhibited pericyte proliferation and migration in vitro. This study highlights the significance of PDGFR-beta signaling for the recruitment, proliferation, and functional activities of fibro-blasts and pericytes during the early phases of wound healing.


Assuntos
Fibroblastos/fisiologia , Pericitos/fisiologia , Piperazinas/farmacologia , Pirimidinas/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Pele/lesões , Cicatrização/efeitos dos fármacos , Animais , Apoptose , Benzamidas , Movimento Celular , Proliferação de Células , Colágeno Tipo I/biossíntese , Colágeno Tipo I/genética , Feminino , Mesilato de Imatinib , Camundongos , Camundongos Transgênicos , Neovascularização Fisiológica/efeitos dos fármacos , Regiões Promotoras Genéticas
12.
Exp Cell Res ; 304(1): 16-27, 2005 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-15707570

RESUMO

Fibroblast proliferation and procollagen production are central features of tissue repair and fibrosis. In addition to its role in blood clotting, the coagulation cascade proteinase thrombin can contribute to tissue repair by stimulating fibroblasts via proteolytic activation of proteinase-activated receptor-1 (PAR1). During hemostasis, the coagulation cascade proteinase factor X is converted into factor Xa. We have previously shown that factor Xa upregulates fibroblast proliferation via production of autocrine PDGF. In this study, we further examined the effects of factor Xa on fibroblast function and aimed to identify its signaling receptor. We showed that factor Xa stimulates procollagen promoter activity and protein production by human and mouse fibroblasts. This effect was independent of PDGF and thrombin production, but dependent on factor Xa proteolytic activity. We also showed that PAR1-deficient mouse fibroblasts did not upregulate procollagen production, mobilize cytosolic calcium, or proliferate in response to factor Xa. Desensitization techniques and PAR1-specific agonists and inhibitors were used to demonstrate that PAR1 mediates factor Xa signaling in human fibroblasts. This is the first report that factor Xa stimulates extracellular matrix production. In contrast with endothelial cells and vascular smooth muscle cells, fibroblasts appear to be the only cell type in which the effects of factor Xa are mediated mainly via PAR1 and not PAR2. These findings are critical for our understanding of tissue repair and fibrotic mechanisms, and for the design of novel approaches to inhibit the profibrotic effects of the coagulation cascade without compromising blood hemostasis.


Assuntos
Sinalização do Cálcio , Fator Xa/fisiologia , Fibroblastos/metabolismo , Pró-Colágeno/biossíntese , Receptor PAR-1/metabolismo , Animais , Proliferação de Células , Fator Xa/genética , Humanos , Pulmão/citologia , Camundongos , Pró-Colágeno/genética , Regiões Promotoras Genéticas
13.
J Biol Chem ; 280(16): 16053-65, 2005 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-15708853

RESUMO

We have generated transgenic mice expressing a kinase-deficient type II transforming growth factor-beta (TGFbeta) receptor selectively on fibroblasts (TbetaRIIDeltak-fib). These mice develop dermal and pulmonary fibrosis. In the present study we explore activation of TGFbeta signaling pathways in this strain and examine the profibrotic properties of explanted transgenic fibroblasts including myofibroblast differentiation and abnormal metalloproteinase production. Gene expression profiles of littermate wild type or transgenic fibroblasts were compared using high-density gene arrays and validated by Taqman reverse transcriptase-PCR, Northern and Western blotting. Using a specific inhibitor (SD-208) we demonstrate that the abnormal phenotype of these cells is dependent upon TbetaRI kinase (ALK5) activity, and that transgenic fibroblasts show enhanced expression and activation of TGFbeta together with increased levels of wild type TbetaRII. Moreover, we confirm that transgene expression is itself regulated by TGFbeta and that expression at low levels facilitates signaling, whereas high level expression is inhibitory. For a subset of TGFbeta responsive genes basal up-regulation is normalized or suppressed by exogenous recombinant TGFbeta1 at time points coincident with increased transgene expression. These findings explain the profound refractoriness of TbetaRIIDeltak-fib fibroblasts to exogenous TGFbeta1, despite their activated phenotype. Thus, transgenic fibroblasts recapitulate many hallmark biochemical properties of fibrotic cells, including high level CTGF (CCN2) expression and type I collagen overproduction, altered MMP production, and myofibroblast differentiation. These cells also show an enhanced ability to contract collagen gel matrices. Our study demonstrates that altered high affinity TGFbeta receptor function may lead to ligand-dependent activation of downstream signaling, and provides further evidence of a pivotal role for sustained TGFbeta overactivity in fibrosis.


Assuntos
Fibroblastos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/genética , Transdução de Sinais/fisiologia , Animais , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/fisiologia , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Transgênicos , Mutação , Fosforilação , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/genética , Proteínas Smad , Transativadores/metabolismo
14.
Am J Pathol ; 162(2): 533-46, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12547711

RESUMO

Transforming growth factor-beta1 (TGF-beta1) plays a central role in promoting extracellular matrix protein deposition by promoting the transformation of fibroblasts to myofibroblasts. To gain new insights into the transcriptional programs involved, we profiled human fetal lung fibroblast global gene expression in response to TGF-beta1 up to 24 hours using oligonucleotide microarrays. In this report, we present data for 146 genes that were up-regulated at least twofold at two time points. These genes group into several major functional categories, including genes involved in cytoskeletal reorganization (n = 30), matrix formation (n = 25), metabolism and protein biosynthesis (n = 27), cell signaling (n = 21), proliferation and survival (n = 13), gene transcription (n = 9), and of uncertain function (n = 21). For 80 of these genes, this is the first report that they are TGF-beta1-responsive. The early induction of two members of the inhibitor of differentiation (ID) family of transcriptional regulators, ID1 and ID3, was followed by the up-regulation of a number of genes that are usually expressed by highly differentiated smooth muscle cells, including smooth muscle myosin heavy chain, basic calponin, and smoothelin. These findings were confirmed at the protein level for primary adult lung fibroblasts. ID1 further behaved like a typical immediate-early gene and, unlike ID3, was expressed and induced at the protein level. Immunohistochemical analysis showed that ID1 was highly expressed by (myo)fibroblasts within fibrotic foci in experimentally induced pulmonary fibrosis. ID1 acts as a dominant-negative antagonist of basic helix-loop-helix transcription factors that drive cell lineage commitment and differentiation. These findings have important implications for our understanding of fibroblast transcriptional programming in response to TGF-beta1 during development, oncogenesis, tissue repair, and fibrosis.


Assuntos
Fibroblastos/fisiologia , Perfilação da Expressão Gênica , Músculo Liso/fisiologia , Proteínas de Neoplasias/genética , Proteínas Repressoras , Transcrição Gênica , Fator de Crescimento Transformador beta/farmacologia , Divisão Celular , Linhagem Celular , Sobrevivência Celular , Feto , Fibroblastos/efeitos dos fármacos , Sequências Hélice-Alça-Hélice , Humanos , Imuno-Histoquímica , Proteína 1 Inibidora de Diferenciação , Proteínas Inibidoras de Diferenciação , Pulmão , Músculo Liso/citologia , Fenótipo , RNA Mensageiro/genética , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA