Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Hepatology ; 61(1): 129-40, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25132354

RESUMEN

UNLABELLED: Alcoholic hepatitis (AH) is a distinct spectrum of alcoholic liver disease (ALD) with intense neutrophilic (polymorphonuclear; PMN) inflammation and high mortality. Although a recent study implicates osteopontin (SPP1) in AH, SPP1 is also shown to have protective effects on experimental ALD. To address this unsettled question, we examined the effects of SPP1 deficiency in male mice given 40% calories derived from ad libitum consumption of the Western diet high in cholesterol and saturated fat and the rest from intragastric feeding of alcohol diet without or with weekly alcohol binge. Weekly binge in this new hybrid feeding model shifts chronic ASH with macrophage inflammation and perisinusoidal and pericellular fibrosis to AH in 57% (15 of 26) of mice, accompanied by inductions of chemokines (Spp1, Cxcl1, and interleukin [Il]-17a), progenitor genes (Cd133, Cd24, Nanog, and epithelial cell adhesion molecule), PMN infiltration, and clinical features of AH, such as hypoalbuminemia, bilirubinemia, and splenomegaly. SPP1 deficiency does not reduce AH incidence and inductions of progenitor and fibrogenic genes, but rather enhances the Il-17a induction and PMN infiltration in some mice. Furthermore, in the absence of SPP1, chronic ASH mice without weekly binge begin to develop AH. CONCLUSION: These results suggest that SPP1 has a protective, rather than causal, role for experimental AH reproduced in our model.


Asunto(s)
Modelos Animales de Enfermedad , Hígado Graso Alcohólico/inmunología , Hepatitis Alcohólica/inmunología , Neutrófilos/fisiología , Osteopontina/metabolismo , Animales , Consumo Excesivo de Bebidas Alcohólicas/complicaciones , Masculino , Ratones Endogámicos C57BL , Receptor Toll-Like 4/metabolismo , alfa-Fetoproteínas/metabolismo
2.
J Biol Chem ; 287(13): 10355-10367, 2012 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-22298767

RESUMEN

Hepatic stellate cells (HSCs) undergo myofibroblastic activation in liver fibrosis and regeneration. This phenotypic switch is mechanistically similar to dedifferentiation of adipocytes as such the necdin-Wnt pathway causes epigenetic repression of the master adipogenic gene Pparγ, to activate HSCs. Now we report that delta-like 1 homolog (DLK1) is expressed selectively in HSCs in the adult rodent liver and induced in liver fibrosis and regeneration. Dlk1 knockdown in activated HSCs, causes suppression of necdin and Wnt, epigenetic derepression of Pparγ, and morphologic and functional reversal to quiescent cells. Hepatic Dlk1 expression is induced 40-fold at 24 h after partial hepatectomy (PH) in mice. HSCs and hepatocytes (HCs) isolated from the regenerating liver show Dlk1 induction in both cell types. In HC and HSC co-culture, increased proliferation and Dlk1 expression by HCs from PH are abrogated with anti-DLK1 antibody (Ab). Dlk1 and Wnt10b expression by Sham HCs are increased by co-culture with PH HSCs, and these effects are abolished with anti-DLK Ab. A tail vein injection of anti-DLK1 Ab at 6 h after PH reduces early HC proliferation and liver growth, accompanied by decreased Wnt10b, nonphosphorylated ß-catenin, p-ß-catenin (Ser-552), cyclins (cyclin D and cyclin A), cyclin-dependent kinases (CDK4, and CDK1/2), p-ERK1/2, and p-AKT. In the mouse developing liver, HSC precursors and HSCs express high levels of Dlk1, concomitant with Dlk1 expression by hepatoblasts. These results suggest novel roles of HSC-derived DLK1 in activating HSCs via epigenetic Pparγ repression and participating in liver regeneration and development in a manner involving the mesenchymal-epithelial interaction.


Asunto(s)
Células Estrelladas Hepáticas/metabolismo , Hepatocitos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Regeneración Hepática , Hígado/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Anticuerpos/farmacología , Proteínas de Unión al Calcio , Línea Celular , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Pollos , Técnicas de Cocultivo , Ciclina A/genética , Ciclina A/metabolismo , Ciclina D/genética , Ciclina D/metabolismo , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Células Estrelladas Hepáticas/patología , Hepatocitos/patología , Péptidos y Proteínas de Señalización Intercelular/genética , Hígado/patología , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Masculino , Proteínas de la Membrana/genética , Ratones , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , PPAR gamma/biosíntesis , PPAR gamma/genética , Proteínas Proto-Oncogénicas c-akt , Ratas , Ratas Wistar , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética
3.
J Gastroenterol Hepatol ; 27 Suppl 2: 94-8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22320925

RESUMEN

Hepatic stellate cells (HSC) are the liver mesenchymal cell type which responds to hepatocellular damage and participates in wound healing. Although HSC myofibroblastic trans-differentiation (activation) is implicated in excessive extracellular matrix deposition, molecular understanding of this phenotypic switch from the viewpoint of cell fate regulation is limited. Recent studies demonstrate the roles of anti-adipogenic morphogens (Wnt, Necdin, Shh) in epigenetic repression of the HSC differentiation gene Pparγ as a causal event in HSC activation. These morphogens have positive cross-interactions which converge to epigenetic repression of Pparγ involving the methyl-CpG binding protein MeCP2. However, these morphogens expressed by activated HSC may also participate in cross-talk between HSC and hepatoblasts/hepatocytes to support liver regeneration, and their aberrant regulation may contribute to liver tumorigenesis. Implications of HSC-derived morphogens in these possibilities are discussed.


Asunto(s)
Células Estrelladas Hepáticas/metabolismo , Hepatopatías/metabolismo , Hígado/metabolismo , Transducción de Señal , Adipogénesis , Animales , Enfermedad Crónica , Epigénesis Genética , Proteínas Hedgehog/metabolismo , Células Estrelladas Hepáticas/patología , Humanos , Hígado/patología , Hepatopatías/genética , Hepatopatías/patología , Hepatopatías/terapia , Morfogénesis , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , PPAR gamma/metabolismo , Fenotipo , Transducción de Señal/genética , Proteínas Wnt/metabolismo
4.
Hepatol Res ; 41(7): 675-82, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21504520

RESUMEN

Research in the past three decades has identified key mediators and signaling mechanisms responsible for myofibroblastic transdifferentiation (MTD) of hepatic stellate cells (HSC), the pivotal event in liver fibrogenesis. Yet, fundamental understanding of the MTD from the viewpoint of cell fate or lineage regulation has been elusive. Recent studies using genetic cell fate mapping techniques demonstrate HSC are derived from mesoderm and at least in part via septum transversum and mesothelium. HSC express markers for different cell types derived from multipotent mesenchymal progenitors. A regulatory commonality between differentiation of adipocytes and that of HSC is shown, and a shift from adipogenic to myogenic or neuronal phenotype characterizes HSC MTD. Central to this shift is a loss of expression of the master adipogenic regulator peroxisome proliferator activated receptor-γ (PPAR-γ). Restored expression of PPAR-γ and/or other adipogenic transcription factors reverses myofibroblastic HSC to differentiated cells. In MTD, Pparγ is epigenetically repressed by induction of methyl-CpG binding protein 2 and its enrichment to the promoter and polycomb repressive complex-facilitated histone H3 lysine 27 di/tri-methylation at the 3' exons. Blocking canonical wingless-related MMTV integration site (Wnt) signaling in myofibroblastic HSC with the co-receptor antagonist Dickkopf-1, abrogates these epigenetic mechanisms, restores PPAR-γ expression and HSC differentiation. Necdin, a melanoma antigen family protein, is identified as an upstream mediator for induction of the canonical Wnt10b and consequent Pparγ repression and HSC MTD. The identified morphogen-induced epigenetic regulation of Pparγ and HSC fate may serve as a novel target for manipulation of liver fibrosis and mesenchymal-epithelial interactions in liver regeneration.

5.
J Biol Chem ; 285(40): 30463-71, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20663865

RESUMEN

Hepatic stellate cells (HSCs), vitamin A-storing liver pericytes, undergo myofibroblastic trans-differentiation or "activation" to participate in liver wound healing. This cellular process involves loss of regulation by adipogenic transcription factors such as peroxisome proliferator-activated receptor γ (PPARγ). Necdin, a melanoma antigen family protein, promotes neuronal and myogenic differentiation while inhibiting adipogenesis. The present study demonstrates that necdin is selectively expressed in HSCs among different liver cell types and induced during their activation in vitro and in vivo. Silencing of necdin with adenovirally expressed shRNA, reverses activated HSCs to quiescent cells in a manner dependent on PPARγ and suppressed canonical Wnt signaling. Promoter analysis, site-directed mutagenesis, and chromatin immunoprecipitation demonstrate that Wnt10b, a canonical Wnt induced in activated HSCs, is a direct target of necdin. Necdin silencing abrogates three epigenetic signatures implicated in repression of PPARγ: increased MeCP2 (methyl CpG binding protein 2) and HP-1α co-repressor recruitments to Pparγ promoter and enhanced H3K27 dimethylation at the exon 5 locus, again in a manner dependent on suppressed canonical Wnt. These epigenetic effects are reproduced by antagonism of canonical Wnt signaling with Dikkopf-1. Our results demonstrate a novel necdin-Wnt pathway, which serves to mediate antiadipogenic HSC trans-differentiation via epigenetic repression of PPARγ.


Asunto(s)
Células Estrelladas Hepáticas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , PPAR gamma/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/genética , Silenciador del Gen , Histonas/genética , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , PPAR gamma/genética , Ratas , Ratas Wistar , Regeneración/efectos de los fármacos , Regeneración/genética , Proteínas Wnt/genética
6.
Gastroenterology ; 138(2): 705-14, 714.e1-4, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19843474

RESUMEN

BACKGROUND & AIMS: Myofibroblast transdifferentiation generates hepatic myofibroblasts, which promote liver fibrogenesis. The peroxisome proliferator-activated receptor gamma (PPARgamma) is a negative regulator of this process. We investigated epigenetic regulation of PPARgamma and myofibroblast transdifferentiation. METHODS: Chromatin immunoprecipitation (ChIP) assays assessed the binding of methyl-CpG binding protein 2 (MeCP2) to PPARgamma and chromatin modifications that silence this gene. MeCP2(-/y) mice and an inhibitor (DZNep) of the epigenetic regulatory protein EZH2 were used in the carbon tetrachloride model of liver fibrosis. Liver tissues from mice were assessed by histologic analysis; markers of fibrosis were measured by quantitative polymerase chain reaction (qPCR). Reverse transcription PCR detected changes in expression of the microRNA miR132 and its target, elongated transcripts of MeCP2. Myofibroblasts were transfected with miR132; PPARgamma and MeCP2 expressions were analyzed by qPCR or immunoblotting. RESULTS: Myofibroblast transdifferentiation of hepatic stellate cells is controlled by a combination of MeCP2, EZH2, and miR132 in a relay pathway. The pathway is activated by down-regulation of miR132, releasing the translational block on MeCP2. MeCP2 is recruited to the 5' end of PPARgamma, where it promotes methylation by H3K9 and recruits the transcription repressor HP1alpha. MeCP2 also stimulates expression of EZH2 and methylation of H3K27 to form a repressive chromatin structure in the 3' exons of PPARgamma. Genetic and pharmacologic disruptions of MeCP2 or EZH2 reduced the fibrogenic characteristics of myofibroblasts and attenuated fibrogenesis. CONCLUSIONS: Liver fibrosis is regulated by an epigenetic relay pathway that includes MeCP2, EZH2, and miR132. Reagents that interfere with this pathway might be developed to reduce fibrogenesis in chronic liver disease.


Asunto(s)
Diferenciación Celular/fisiología , Epigénesis Genética/fisiología , Fibroblastos/fisiología , Cirrosis Hepática/fisiopatología , Proteína 2 de Unión a Metil-CpG/fisiología , Transducción de Señal/fisiología , Animales , Tetracloruro de Carbono/efectos adversos , Transdiferenciación Celular , Modelos Animales de Enfermedad , Proteína Potenciadora del Homólogo Zeste 2 , Células Estrelladas Hepáticas/fisiología , N-Metiltransferasa de Histona-Lisina/fisiología , Cirrosis Hepática/inducido químicamente , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/fisiología , PPAR gamma/fisiología , Complejo Represivo Polycomb 2
7.
Nucleic Acids Res ; 36(1): 179-88, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18003659

RESUMEN

Mechanisms of gene repression by transforming growth factor-beta (TGF-beta) are not well understood. TGF-beta represses transcription of pulmonary surfactant protein-B gene in lung epithelial cells. Repression is mediated by SMAD3 through interactions with NKX2.1 and FOXA1, two key transcription factors that are positive regulators of SpB transcription. In this study, we found that SMAD3 interacts through its MAD domains, MH1 and MH2 with NKX2.1 and FOXA1 proteins. The sites of interaction on NKX2.1 are located within the NH2 and COOH domains, known to be involved in transactivation function. In comparison, weaker interaction of FOXA1 winged helix, and the NH(2)-terminal domains was documented with SMAD3. Both in vitro studies and in vivo ChIP assays show that interaction of SMAD3 MH1 and MH2 domains with NKX2.1 and FOXA1 results in reduced binding of NKX2.1 and FOXA1 to their cognate DNA-binding sites, and diminished promoter occupancy within the SpB promoter. Thus, these studies reveal for the first time a mechanism of TGF-beta-induced SpB gene repression that involves interactions between specific SMAD3 domains and the corresponding functional sites on NKX2.1 and FOXA1 transcription factors.


Asunto(s)
Factor Nuclear 3-alfa del Hepatocito/metabolismo , Proteínas Nucleares/metabolismo , Proteína B Asociada a Surfactante Pulmonar/genética , Proteínas Represoras/química , Proteína smad3/química , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Línea Celular , Factor Nuclear 3-alfa del Hepatocito/química , Humanos , Inmunoprecipitación , Proteínas Nucleares/química , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Ratas , Proteínas Represoras/metabolismo , Proteína smad3/metabolismo , Factor Nuclear Tiroideo 1 , Factores de Transcripción/química , Factor de Crecimiento Transformador beta/farmacología , Técnicas del Sistema de Dos Híbridos
8.
Dev Dyn ; 237(2): 328-38, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18161062

RESUMEN

Lung maturation is hallmarked by the appearance of surfactant-producing alveoli during transition from the saccular to alveolar stage of lung development. Inflammation can disrupt this process and accelerate lung maturity following intrauterine amniotic infection (chorioamnionitis). Nuclear factor kB (NF-kB) is a transcription factor central to multiple inflammatory and developmental pathways, including dorsal-ventral patterning in fruit flies, limb and mammary and submandibular gland development in mice, and branching morphogenesis in chick lungs. Given its shared role in inflammation and developmental signaling, we hypothesized that overexpression of NF-kB targeted to the lung epithelium would exert maturational effects on alveolar development. We generated transgenic mice with lung-specific overexpression of the RelA subunit of NF-kB using a surfactant protein C promoter construct. Our results showed that RelA overexpression in the lung yields increased alveolar type I and type II cells. These findings are consistent with a model whereby NF-kB may induce maturation of lung development through decreased apoptosis of epithelial cells.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Pulmón/embriología , Morfogénesis/fisiología , Factor de Transcripción ReIA/metabolismo , Animales , Apoptosis/fisiología , Acuaporina 5/metabolismo , Northern Blotting , Western Blotting , Bromodesoxiuridina , Inmunohistoquímica , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Péptidos y Proteínas de Señalización Intercelular , Pulmón/ultraestructura , Ratones , Ratones Transgénicos , Microscopía Electrónica de Rastreo , Proteínas Nucleares/metabolismo , Péptidos/metabolismo , Regiones Promotoras Genéticas/genética , Proteína C/genética , Proteína C Asociada a Surfactante Pulmonar , Factor Nuclear Tiroideo 1 , Factores de Transcripción/metabolismo
9.
Gene ; 393(1-2): 70-80, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17350185

RESUMEN

Bone Morphogenetic Proteins are key signaling molecules in vertebrate development. Little is known about Bmp gene regulation in any organ. In Drosophila, the Bmp gene, dpp is regulated by Dorsal, the invertebrate homologue of Rel-NF-kB. In this study we examined whether TNF-alpha, which activates NF-kB, can regulate Bmp4 gene expression. TNF-alpha reduced Bmp4 mRNA in lung adenocarcinoma A549 cells and repressed transcriptional activity of the human Bmp4 promoter in a dose-dependent manner. Similar repression was observed when the Bmp4 promoter was co-transfected with a p65 (RelA) expression vector in the absence of TNF-alpha treatment, suggesting that RelA mediates the effect of TNF-alpha. In support of this finding, the repressor effect of TNF-alpha on Bmp4 was abrogated by a co-transfected dominant negative mutant of IkB (S32A/S36A). The human Bmp4 promoter contains 3 putative consensus binding sites for NF-kB. Surprisingly, only one of the latter binding sites was capable of binding NF-kB. Repressor effect of NF-kB was not dependent on any of the three binding sites, but localized to a 122 bp fragment which bound both RelA and SP1. SP1 stimulated transcription, whereas increasing doses of RelA opposed this effect. In vivo, TNF-alpha inhibited branching morphogenesis and LacZ gene expression in Bmp4-lacz transgenic lungs. These data support a model in which TNF-alpha-induced RelA interacts with SP1 to bring about transcriptional repression of Bmp4 gene. These findings provide a mechanistic paradigm for interactions between mediators of inflammation and morphogenesis with relevant implications for normal lung development and pathogenesis of disease.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Pulmón/citología , Pulmón/metabolismo , Transcripción Genética/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Animales , Emparejamiento Base/genética , Secuencia de Bases , Sitios de Unión , Proteína Morfogenética Ósea 4 , Proteínas Morfogenéticas Óseas/metabolismo , Línea Celular Tumoral , Elementos de Facilitación Genéticos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Pulmón/efectos de los fármacos , Pulmón/embriología , Ratones , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Eliminación de Secuencia , Factor de Transcripción ReIA/metabolismo
10.
J Neurosurg ; 103(6): 1058-66, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16381193

RESUMEN

OBJECT: Adenovirus vector (AdV)-mediated gene delivery has been recently demonstrated in clinical trials as a novel potential treatment for malignant gliomas. Combined coxsackievirus B and adenovirus receptor (CAR) has been shown to function as an attachment receptor for multiple adenovirus serotypes, whereas the vitronectin integrins (alphavbeta3 and alphavbeta5) are involved in AdV internalization. In resected glioma specimens, the authors demonstrated that malignant gliomas have varying levels of CAR, alphavbeta3, and alphavbeta5 expression. METHODS: A correlation between CAR expression and the transduction efficiency of AdV carrying the green fluorescent protein in various human glioblastoma multiforme (GBM) cell lines and GBM primary cell lines was observed. To increase transgene activity in in vitro glioma cells with low or deficient levels of CAR, the authors used basic fibroblast growth factor (FGF2) as a targeting ligand to redirect adenoviral infection through its cognate receptor, FGF receptor 1 (FGFR1), which was expressed at high levels by all glioma cells. These findings were confirmed by in vivo study data demonstrating enhanced transduction efficiency of FGF2-retargeted AdV in CAR-negative intracranial gliomas compared with AdV alone, without evidence of increased angiogenesis. CONCLUSIONS: Altogether, the results demonstrated that AdV-mediated gene transfer using the FGF2/FGFR system is effective in gliomas with low or deficient levels of CAR and suggested that FGF2-retargeting of AdV may be a promising approach in glioma gene therapy.


Asunto(s)
Adenoviridae/genética , Factor 2 de Crecimiento de Fibroblastos/genética , Marcación de Gen , Terapia Genética , Vectores Genéticos , Glioma/terapia , Línea Celular Tumoral , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Humanos , Integrina alfaVbeta3/metabolismo , Integrinas/metabolismo , Ligandos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptores Virales/metabolismo , Receptores de Vitronectina/metabolismo , Transducción Genética
11.
Dev Dyn ; 228(1): 82-94, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12950082

RESUMEN

The Clock gene, timeless, regulates circadian rhythm in Drosophila, but its vertebrate homolog is critical to embryonic development. Timeless was shown to be involved in murine urethral bud branching morphogenesis. We generated a polyclonal antibody to mouse TIMELESS (mTIM) and studied its distribution and its potential role during lung development, which also requires branching morphogenesis. In the early mouse embryo, TIM was localized to all organs, especially the neural epithelium. In embryonic day (E) 9.5 embryos, TIM was present in both epithelial and mesenchymal cells at the onset of lung morphogenesis. In E15 embryos, TIM decreased in the mesenchyme but remained pronounced in the epithelium of both large and small airways. Later, TIM was localized to a specific subset of epithelial cells with alveolar type 2 phenotype. This finding was verified by immunostaining of isolated alveolar type 2 cells. In the proximal airways, TIM was colocalized with CCSP to nonciliated columnar epithelial cells. Antisense oligonucleotides to mTim specifically inhibited branching morphogenesis of embryonic lungs in explant culture without affecting SpC expression an alveolar type 2 cell marker. In cultured lung cells, expression of TIM is independent of cell cycle and proliferation. These studies indicate that the function of Timeless is highly conserved in organs whose formation requires branching morphogenesis.


Asunto(s)
Pulmón/embriología , Morfogénesis , Factores de Transcripción/metabolismo , Células 3T3 , Animales , Biomarcadores , Proteínas de Ciclo Celular , Diferenciación Celular , Ritmo Circadiano , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células HeLa , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular , Pulmón/citología , Pulmón/efectos de los fármacos , Mesodermo/citología , Mesodermo/metabolismo , Ratones/embriología , Oligonucleótidos Antisentido/farmacología , Técnicas de Cultivo de Órganos , Factores de Tiempo
12.
J Biol Chem ; 277(41): 38399-408, 2002 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-12161428

RESUMEN

Transforming growth factor-beta (TGF-beta) represses surfactant protein B (Sp-B) gene transcription through a mechanism that remains unknown. A homeodomain and a forkhead transcription factor, NKX2.1 and HNF-3, respectively, are known activators of Sp-B transcription. Because SMADs are the effectors of TGF-beta-induced gene activation, we examined the possibility that gene repression by TGF-beta may also occur through interactions of SMADs with NKX2.1 and HNF-3. We found that lung epithelial carcinoma H441 cells contain SMAD2/3 and -4, which localize to the nucleus in response to TGF-beta treatment. The activity of a transfected Sp-B promoter/reporter construct was reduced in a dose-dependent manner by TGF-beta. Cotransfection with a mutant, constitutively activated form of the Tgf-beta type I receptor repressed Sp-B promoter activity in the absence of TGF-beta ligand. Dominant negative mutants of Smads blocked the repressor activity of TGF-beta. SMAD3, but not SMAD2, mediated the repressor activity of TGF-beta on the Sp-B promoter. Mutations within a 70-base pair domain that includes binding sites for NKX2.1, hepatocyte nuclear factor 3 (HNF-3), or cAMP response element-binding protein (CREB) eliminated SMAD3-dependent repression of Sp-B transcription. Electrophoretic mobility shift analysis showed no evidence for direct binding of SMAD3 to the Sp-B promoter, and a DNA binding mutant of SMAD3 also repressed Sp-B, suggesting that direct DNA binding of SMAD3 may not be required. Using a mammalian two hybrid assay, we found physical and functional interactions between SMAD3 and both NKX2.1 and HNF-3. Also, a glutathione S-transferase-fused SMAD3 directly binds to in vitro synthesized NKX2.1 or HNF-3, demonstrating protein-protein interactions between SMAD3 and the two transcriptional factors. The DNA binding of NKX2.1 to Sp-B promoter was reduced in response to TGF-beta treatment, although expression of Nkx2.1 was not affected. We conclude that SMAD3 interactions with the positive regulators NKX2.1 and HNF-3 underlie the molecular basis for TGF-beta-induced repression of Sp-B gene transcription.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Proteínas Nucleares/metabolismo , Proteína B Asociada a Surfactante Pulmonar/genética , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Factor de Crecimiento Transformador beta/metabolismo , Receptores de Activinas Tipo I/metabolismo , Carcinoma , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Células Epiteliales/citología , Genes Reporteros , Glutatión Transferasa/metabolismo , Humanos , Neoplasias Pulmonares , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Serina-Treonina Quinasas , Proteína B Asociada a Surfactante Pulmonar/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Proteína smad3 , Factor Nuclear Tiroideo 1 , Activación Transcripcional , Factor de Crecimiento Transformador beta1 , Células Tumorales Cultivadas , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA