Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
PLoS One ; 14(11): e0225422, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31765403

RESUMEN

Type 2 insulin-like growth factor (IGF-II) levels are increased in fibrosing lung diseases such as idiopathic pulmonary fibrosis (IPF) and scleroderma/systemic sclerosis-associated pulmonary fibrosis (SSc). Our goal was to investigate the contribution of IGF receptors to IGF-II-mediated fibrosis in these diseases and identify other potential mechanisms key to the fibrotic process. Cognate receptor gene and protein expression were analyzed with qRT-PCR and immunoblot in primary fibroblasts derived from lung tissues of normal donors (NL) and patients with IPF or SSc. Compared to NL, steady-state receptor gene expression was decreased in SSc but not in IPF. IGF-II stimulation differentially decreased receptor mRNA and protein levels in NL, IPF, and SSc fibroblasts. Neutralizing antibody, siRNA, and receptor inhibition targeting endogenous IGF-II and its primary receptors, type 1 IGF receptor (IGF1R), IGF2R, and insulin receptor (IR) resulted in loss of the IGF-II response. IGF-II tipped the TIMP:MMP balance, promoting a fibrotic environment both intracellularly and extracellularly. Differentiation of fibroblasts into myofibroblasts by IGF-II was blocked with a TGFß1 receptor inhibitor. IGF-II also increased TGFß2 and TGFß3 expression, with subsequent activation of canonical SMAD2/3 signaling. Therefore, IGF-II promoted fibrosis through IGF1R, IR, and IGF1R/IR, differentiated fibroblasts into myofibroblasts, decreased protease production and extracellular matrix degradation, and stimulated expression of two TGFß isoforms, suggesting that IGF-II exerts pro-fibrotic effects via multiple mechanisms.


Asunto(s)
Fibroblastos/metabolismo , Factor II del Crecimiento Similar a la Insulina/metabolismo , Fibrosis Pulmonar/metabolismo , Células Cultivadas , Fibroblastos/patología , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Fibrosis Pulmonar/patología , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Transducción de Señal , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
2.
Arthritis Rheum ; 63(3): 783-94, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21360508

RESUMEN

OBJECTIVE: Pulmonary complications, including pulmonary fibrosis (PF) and pulmonary arterial hypertension (PAH), are the leading cause of mortality in patients with systemic sclerosis (SSc). The aim of this study was to compare the molecular fingerprint of lung tissue and matching primary fibroblasts from patients with SSc with that of lung tissue and fibroblasts from normal donors, patients with idiopathic pulmonary fibrosis (IPF), and patients with idiopathic pulmonary arterial hypertension (IPAH). METHODS: Lung tissue samples were obtained from 33 patients with SSc who underwent lung transplantation. Tissues and cells from a subgroup of SSc patients with predominantly PF or PAH were compared to those from normal donors, patients with IPF, and patients with IPAH. Microarray data were analyzed using efficiency analysis for determination of the optimal data-processing methods. Real-time polymerase chain reaction and immunohistochemistry were used to confirm differential levels of messenger RNA and protein, respectively. RESULTS: Consensus efficiency analysis identified 242 and 335 genes that were differentially expressed in lungs and primary fibroblasts, respectively. SSc-PF and IPF lungs shared enriched functional groups in genes implicated in fibrosis, insulin-like growth factor signaling, and caveolin-mediated endocytosis. Gene functional groups shared by SSc-PAH and IPAH lungs included those involved in antigen presentation, chemokine activity, and interleukin-17 signaling. CONCLUSION: Using microarray analysis on carefully phenotyped SSc and comparator lung tissues, we demonstrated distinct molecular profiles in tissues and fibroblasts from patients with SSc-associated lung disease compared to idiopathic forms of lung disease. Unique molecular signatures were generated that are disease specific (SSc) and phenotype specific (PF versus PAH). These signatures provide new insights into the pathogenesis and potential therapeutic targets of SSc-related lung disease.


Asunto(s)
Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/patología , Adulto , Western Blotting , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Hipertensión Pulmonar/cirugía , Pulmón/metabolismo , Pulmón/patología , Trasplante de Pulmón , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Fibrosis Pulmonar/cirugía , ARN Mensajero/metabolismo , Esclerodermia Sistémica/cirugía , Transcriptoma
3.
Curr Rheumatol Rep ; 12(1): 19-25, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20425529

RESUMEN

Interstitial lung disease is a frequent complication of systemic sclerosis and currently is the leading cause of death. Our ability to predict which individuals are at greatest risk of developing clinically significant, progressive interstitial lung disease remains inadequate. Identification of circulating autoantibodies and other biomarkers, as well as genetic polymorphisms and aberrant gene expression, all hold promise as diagnostic and prognostic tools, as well as therapeutic targets. Many practice patterns for the diagnosis and monitoring of connective tissue disease-associated interstitial lung disease are based upon published experience with idiopathic interstitial lung diseases. Although there are likely commonalities in the pathophysiologic mechanisms and clinical progression among all fibrosing lung diseases, a better understanding of features unique to systemic sclerosis-associated interstitial lung disease is essential to the development of more effective monitoring and treatment strategies.


Asunto(s)
Fibrosis Pulmonar Idiopática/etiología , Fibrosis Pulmonar Idiopática/fisiopatología , Esclerodermia Sistémica/complicaciones , Epigénesis Genética , Humanos , Fibrosis Pulmonar Idiopática/genética , Esclerodermia Sistémica/genética
4.
Am J Pathol ; 175(2): 605-15, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19628764

RESUMEN

We have previously shown that insulin-like growth factor (IGF) binding protein- 5 (IGFBP-5) is overexpressed in lung fibrosis and induces the production of extracellular matrix components, such as collagen and fibronectin, both in vitro and in vivo. The exact mechanism by which IGFBP-5 exerts these novel fibrotic effects is unknown. We thus examined the signaling cascades that mediate IGFBP-5-induced fibrosis. We demonstrate for the first time that IGFBP-5 induction of extracellular matrix occurs independently of IGF-I, and results from IGFBP-5 activation of MAPK signaling, which facilitates the translocation of IGFBP-5 to the nucleus. We examined the effects of IGFBP-5 on early growth response (Egr)-1, a transcription factor that is central to growth factor-mediated fibrosis. Egr-1 was up-regulated by IGFBP-5 in a MAPK-dependent manner and bound to nuclear IGFBP-5. In fibroblasts from Egr-1 knockout mice, induction of fibronectin by IGFBP-5 was abolished. Expression of Egr-1 in these cells rescued the extracellular matrix-promoting effects of IGFBP-5. Moreover, IGFBP-5 induced cell migration in an Egr-1-dependent manner. Notably, Egr-1 levels, similar to IGFBP-5, were increased in vivo in lung tissues and in vitro in primary fibroblasts of patients with pulmonary idiopathic fibrosis. Taken together, our findings suggest that IGFBP-5 induces a fibrotic phenotype via the activation of MAPK signaling and the induction of nuclear Egr-1 that interacts with IGFBP-5 and promotes fibrotic gene transcription.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fibrosis Pulmonar/metabolismo , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Movimiento Celular , Núcleo Celular/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Activación Enzimática , Fibronectinas/biosíntesis , Humanos , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Ratones , Ratones Noqueados , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Transcripción Genética
5.
Am J Pathol ; 172(6): 1580-90, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18467708

RESUMEN

Systemic sclerosis (SSc)-related pulmonary fibrosis, for which there are few effective therapies, is the most common cause of SSc-related mortality. We examined insulin-like growth factor (IGF)-II expression in explanted lung tissues from control and SSc patients to determine its role in the pathogenesis of fibrosis. IGF-II levels in vivo were detected using immunohistochemistry. Primary lung fibroblasts were cultured from lung tissues, and IGF-II mRNA was measured using reverse transcriptase-polymerase chain reaction. Western blot analysis measured extracellular matrix (ECM) production and phosphorylated signaling molecules. Immunostaining revealed increased IGF-II expression in fibroblastic foci of SSc lungs. Furthermore, primary SSc lung fibroblasts had a fourfold increase in IGF-II mRNA and a twofold increase in IGF-II protein compared with normal lung fibroblasts. IGF-II mRNA in SSc lung fibroblasts was expressed primarily from the P3 promoter of the IGF-II gene, and IGF-II induced both a dose- and time-dependent increase in collagen type I and fibronectin production. IGF-II triggered the activation of both phosphatidylinositol-3 kinase and Jun N-terminal kinase signaling cascades, the inhibition of which diminished IGF-II-induced ECM production. Our study demonstrates increased local IGF-II expression in SSc-associated pulmonary fibrosis both in vitro and in vivo as well as IGF-II-induced ECM production through both phosphatidylinositol-3 kinase- and Jun N-terminal kinase-dependent pathways. Our results provide novel insights into the role of IGF-II in the pathogenesis of SSc-associated pulmonary fibrosis.


Asunto(s)
Factor II del Crecimiento Similar a la Insulina/fisiología , MAP Quinasa Quinasa 4/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fibrosis Pulmonar/metabolismo , Esclerodermia Sistémica/metabolismo , Adulto , Células Cultivadas , Activación Enzimática , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Pulmón/metabolismo , Pulmón/patología , Masculino , Persona de Mediana Edad , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/patología , Esclerodermia Sistémica/complicaciones , Esclerodermia Sistémica/patología , Transducción de Señal/fisiología
6.
Crit Rev Eukaryot Gene Expr ; 18(1): 47-56, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18197785

RESUMEN

Pulmonary fibrosis is a phenotype that results from a variety of conditions and is associated with significant morbidity and mortality. Ongoing research in the field is driven by the need for effective treatments for pulmonary fibrosis. In this review, we highlight mechanisms that regulate gene expression in pulmonary fibrosis at multiple levels. Potential pathogenic mechanisms involve genetic background and transcriptional, posttranscriptional, translational, posttranslational, and epigenetic mechanisms. Pulmonary fibrosis results from abnormal gene expression and regulation that arise from a combination of inherited/acquired genetic alterations and environmental triggers. Collectively, these alterations result in increased expression of extracellular matrix components such as collagen and fibronectin and in the observed fibrosis. Insights gained from mechanisms identified to induce and/or perpetuate fibrosis in the lung will yield new targets for the development of more effective therapies.


Asunto(s)
Expresión Génica , Fibrosis Pulmonar/genética , Animales , Epigénesis Genética , Humanos , Modelos Biológicos , Procesamiento Proteico-Postraduccional , Fibrosis Pulmonar/metabolismo , Interferencia de ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA