Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Biol Chem ; 288(48): 34757-66, 2013 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-24129565

RESUMEN

Inflammatory immune responses play an important role in mucosal homeostasis and gut diseases. Nuclear factor κB (NF-κB), central to the proinflammatory cascade, is activated in necrotizing enterocolitis (NEC), a devastating condition of intestinal injury with extensive inflammation in premature infants. TGF-ß is a strong immune suppressor and a factor in breast milk, which has been shown to be protective against NEC. In an NEC animal model, oral administration of the isoform TGF-ß1 activated the downstream effector Smad2 in intestine and significantly reduced NEC incidence. In addition, TGF-ß1 suppressed NF-κB activation, maintained levels of the NF-κB inhibitor IκBα in the intestinal epithelium, and systemically decreased serum levels of IL-6 and IFN-γ. The immature human fetal intestinal epithelial cell line H4 was used as a reductionistic model of the immature enterocyte to investigate mechanism. TGF-ß1 pretreatment inhibited the TNF-α-induced IκBα phosphorylation that targets the IκBα protein for degradation and inhibited NF-κB activation. Chromatin immunoprecipitation (ChIP) assays demonstrated decreased NF-κB binding to the promoters of IL-6, IL-8, and IκBα in response to TNF-α with TGF-ß1 pretreatment. These TGF-ß1 effects appear to be mediated through the canonical Smad pathway as silencing of the TGF-ß central mediator Smad4 resulted in loss of the TGF-ß1 effects. Thus, TGF-ß1 is capable of eliciting anti-inflammatory effects by inhibiting NF-κB specifically in the intestinal epithelium as well as by decreasing systemic IL-6 and IFN-γ levels. Oral administration of TGF-ß1 therefore can potentially be used to protect against gastrointestinal diseases.


Asunto(s)
Enfermedades Gastrointestinales/tratamiento farmacológico , Inflamación/metabolismo , FN-kappa B/metabolismo , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/administración & dosificación , Administración Oral , Animales , Línea Celular , Enfermedades Gastrointestinales/metabolismo , Enfermedades Gastrointestinales/patología , Humanos , Inmunidad Innata/genética , Inflamación/tratamiento farmacológico , Inflamación/genética , Interferón gamma/metabolismo , Interleucina-6/metabolismo , Interleucina-8 , Mucosa Intestinal/lesiones , Mucosa Intestinal/metabolismo , Regiones Promotoras Genéticas , Ratas , Transducción de Señal , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
2.
NMR Biomed ; 27(3): 272-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24318809

RESUMEN

Neonatal necrotizing enterocolitis (NEC) is a poorly understood life-threatening illness afflicting premature infants. Research is hampered by the absence of a suitable method to monitor disease progression noninvasively. The primary goal of this research was to test in vivo MRI methods for the noninvasive early detection and staging of inflammation in the ileum of an infant rat model of NEC. Neonatal rats were delivered by cesarean section at embryonic stage of day 20 after the beginning of pregnancy and stressed with formula feeding, hypoxia and bacterial colonization to induce NEC. Naturally born and dam-fed neonatal rats were used as healthy controls. In vivo MRI studies were performed using a Bruker 9.4-T scanner to obtain high-resolution anatomical MR images using both gradient echo and spin echo sequences, pixel-by-pixel T2 maps using a multi-slice-multi-echo sequence, and maps of the apparent diffusion coefficient (ADC) of water using a spin echo sequence, to assess the degree of ileal damage. Pups were sacrificed at the end of the MRI experiment on day 2 or 4 for histology. T2 measured by MRI was increased significantly in the ileal regions of pups with NEC by histology (106.3 ± 6.1 ms) compared with experimentally stressed pups without NEC (85.2 ± 6.8 ms) and nonstressed, control rat pups (64.9 ± 2.3 ms). ADC values measured by diffusion-weighted MRI were also increased in the ileal regions of pups with NEC by histology [(1.98 ± 0.15) × 10(-3) mm(2)/s] compared with experimentally stressed pups without NEC [(1.43 ± 0.16) × 10(-3) mm(2)/s] and nonstressed control pups [(1.10 ± 0.06) × 10(-3) mm(2)/s]. Both T2 and ADC values between these groups were found to be significantly different (p < 0.03). The correlation of MRI results with histologic images of the excised ileal tissue samples strongly suggests that MRI can noninvasively identify NEC and assess intestinal injury prior to clinical symptoms in a physiologic rat pup model of NEC.


Asunto(s)
Enterocolitis Necrotizante/diagnóstico , Imagen por Resonancia Magnética , Animales , Difusión , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/patología , Femenino , Intestinos/patología , Embarazo , Ratas , Ratas Sprague-Dawley , Marcadores de Spin
3.
J Biol Chem ; 286(14): 12123-32, 2011 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-21262973

RESUMEN

The impermeant nature of the intestinal barrier is maintained by tight junctions (TJs) formed between adjacent intestinal epithelial cells. Disruption of TJs and loss of barrier function are associated with a number of gastrointestinal diseases, including neonatal necrotizing enterocolitis (NEC), the leading cause of death from gastrointestinal diseases in preterm infants. Human milk is protective against NEC, and the human milk factor erythropoietin (Epo) has been shown to protect endothelial cell-cell and blood-brain barriers. We hypothesized that Epo may also protect intestinal epithelial barriers, thereby lowering the incidence of NEC. Our data demonstrate that Epo protects enterocyte barrier function by supporting expression of the TJ protein ZO-1. As immaturity is a key factor in NEC, Epo regulation of ZO-1 in the human fetal immature H4 intestinal epithelial cell line was examined and demonstrated Epo-stimulated ZO-1 expression in a dose-dependent manner through the PI3K/Akt pathway. In a rat NEC model, oral administration of Epo lowered the incidence of NEC from 45 to 23% with statistical significance. In addition, Epo treatment protected intestinal barrier function and prevented loss of ZO-1 at the TJs in vivo. These effects were associated with elevated Akt phosphorylation in the intestine. This study reveals a novel role of Epo in the regulation of intestinal epithelial TJs and barrier function and suggests the possible use of enteral Epo as a therapeutic agent for gut diseases.


Asunto(s)
Enterocolitis Necrotizante/tratamiento farmacológico , Eritropoyetina/farmacología , Mucosa Intestinal/metabolismo , Intestinos/citología , Intestinos/efectos de los fármacos , Animales , Animales Recién Nacidos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Impedancia Eléctrica , Enterocitos/citología , Enterocitos/efectos de los fármacos , Enterocitos/metabolismo , Eritropoyetina/uso terapéutico , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Mucosa Intestinal/citología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Ratas , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-1
4.
Pediatr Res ; 69(5 Pt 1): 395-400, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21263375

RESUMEN

Preterm infants face many challenges in transitioning from the in utero to extrauterine environment while still immature. Failure of the preterm gut to successfully mature to accommodate bacteria and food substrate leads to significant morbidity such as neonatal necrotizing enterocolitis. The intestinal epithelial barrier plays a critical role in gut protection. Heat shock protein 70 (Hsp70) is an inducible cytoprotective molecule shown to protect the intestinal epithelium in adult models. To investigate the hypothesis that Hsp70 may be important for early protection of the immature intestine, Hsp70 expression was evaluated in intestine of immature rat pups. Data demonstrate that Hsp70 is induced by exposure to mother's milk. Hsp70 is found in mother's milk, and increased Hsp70 transcription is induced by mother's milk. This Hsp70 colocalizes with the tight junction protein ZO-1. Mother's milk-induced Hsp70 may contribute to maintenance of barrier function in the face of oxidant stress. Further understanding of the means by which mother's milk increases Hsp70 in the ileum will allow potential means of strengthening the intestinal barrier in at-risk preterm infants.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Íleon/metabolismo , Mucosa Intestinal/metabolismo , Leche/metabolismo , Animales , Animales Recién Nacidos , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/microbiología , Citoprotección , Modelos Animales de Enfermedad , Proteínas HSP70 de Choque Térmico/genética , Hipoxia/metabolismo , Íleon/microbiología , Íleon/patología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Proteínas de la Membrana/metabolismo , Estrés Oxidativo , Permeabilidad , Fosfoproteínas/metabolismo , ARN Mensajero/metabolismo , Ratas , Uniones Estrechas/metabolismo , Regulación hacia Arriba , Proteína de la Zonula Occludens-1
5.
Cancer Res ; 67(4): 1571-9, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17308096

RESUMEN

We have recently reported that the expression of a tight junction protein, claudin-1, is increased during colon carcinogenesis and particularly metastatic colorectal cancer. Manipulation of claudin-1 levels in colon cancer cells showed a positive correlation between claudin-1 expression and tumor growth and metastasis. However, the mechanisms underlying the increased claudin-1 expression in colorectal cancer remains unknown. The tumor suppressor Smad4 is a central intracellular signal transduction component of the transforming growth factor-beta (TGF-beta) family of cytokines. Loss of Smad4 protein expression is correlated with poor prognosis and is frequently observed in invasive and metastatic colorectal carcinoma. In the present study, we report an inverse relationship between Smad4 and claudin-1 expression in human colorectal carcinoma tumor samples and in human colon cancer cell lines. We found that the expression of Smad4 in Smad4-deficient but claudin-1-positive SW480 or HT29 colon cancer cell lines down-regulates claudin-1 expression through transcriptional repression by modulating beta-catenin/T-cell factor/lymphocyte enhancer factor activity. Furthermore, this Smad4-dependent inhibition of claudin-1 expression is independent of TGF-beta signaling because Smad4 expression alone is insufficient to restore TGF-beta signaling in the SW480 cells, and the selective TGF-beta receptor kinase inhibitor LY364947 did not prevent the Smad4 suppression of claudin-1 protein expression in either SW480 or HT29 cells. Taken together, these findings suggest a novel mechanism underlying Smad4 tumor-suppressive function through regulation of a potential metastatic modulator, claudin-1, in a TGF-beta-independent manner.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Proteínas de la Membrana/biosíntesis , Proteína Smad4/biosíntesis , Animales , Línea Celular Tumoral , Claudina-1 , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Células HCT116 , Células HT29 , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Invasividad Neoplásica , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Transducción de Señal , Proteína Smad4/deficiencia , Factores de Transcripción TCF/metabolismo , Transcripción Genética , Transfección , Factor de Crecimiento Transformador beta/metabolismo , beta Catenina/metabolismo
6.
J Clin Invest ; 115(7): 1765-76, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15965503

RESUMEN

Disruption of the cell-cell junction with concomitant changes in the expression of junctional proteins is a hallmark of cancer cell invasion and metastasis. The role of adherent junction proteins has been studied extensively in cancer, but the roles of tight junction (TJ) proteins are less well understood. Claudins are recently identified members of the tetraspanin family of proteins, which are integral to the structure and function of TJs. Recent studies show changes in expression/cellular localization of claudins during tumorigenesis; however, a causal relationship between claudin expression/localization and cancer has not been established. Here, we report an increased expression of claudin-1 in human primary colon carcinoma and metastasis and in cell lines derived from primary and metastatic tumors. We also report frequent nuclear localization of claudin-1 in these samples. Genetic manipulations of claudin-1 expression in colon cancer cell lines induced changes in cellular phenotype, with structural and functional changes in markers of epithelial-mesenchymal transition. Furthermore, we demonstrate that changes in claudin-1 expression have significant effects on growth of xenografted tumors and metastasis in athymic mice. We further provide data suggesting that the regulation of E-cadherin expression and beta-catenin/Tcf signaling is a possible mechanism underlying claudin-1-dependent changes.


Asunto(s)
Neoplasias del Colon/etiología , Proteínas de la Membrana/fisiología , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Claudina-1 , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Neoplasias del Colon/secundario , Proteínas del Citoesqueleto/metabolismo , Femenino , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Metástasis de la Neoplasia/fisiopatología , Trasplante de Neoplasias , ARN Interferente Pequeño/genética , Transducción de Señal , Transactivadores/metabolismo , Trasplante Heterólogo , beta Catenina
7.
Cancer Gene Ther ; 11(2): 135-47, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14647233

RESUMEN

To improve the efficacy of tumor cell-based and dendritic cell (DC)-based cancer vaccines, this study explored the potential of a new cancer vaccine strategy, that is, the use of CD40 ligand-transfected tumor (CD40L-tumor) cells to simultaneously deliver both tumor-derived antigens (Ag) and maturation stimuli to DCs. Materials from frozen/thawed or irradiated human tumor cells, with or without surface CD40L, were internalized efficiently by immature DCs after coincubation. However, during the internalization process, only coculturing with irradiated CD40L-tumor cells resulted in concurrent, optimal DC maturation and production of proinflammatory chemokines and pro-Th1 cytokines, such as IL-6, IL-8, IL-12, IFN-gamma, and TNF-alpha. These activated DCs were the most potent cells to support the growth of CD8+, IFN-gamma-producing T cells, and to process tumor Ag for the generation of specific cytotoxic T cells in vitro. Animals vaccinated with irradiated CD40L-tumor cell-pulsed DCs were better protected against subsequent challenge of a weakly immunogenic tumor cell line than animals vaccinated with irradiated CD40L-tumor cells alone. Thus, our results strongly support the future clinical application of using DCs pulsed with irradiated CD40L-tumor cells as a cancer vaccine.


Asunto(s)
Antígenos de Neoplasias/inmunología , Ligando de CD40/genética , Vacunas contra el Cáncer , Células Dendríticas/inmunología , Neoplasias Experimentales/prevención & control , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos CD , Ligando de CD40/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo , Citocinas/metabolismo , Células Dendríticas/trasplante , Humanos , Inmunoglobulinas/metabolismo , Activación de Linfocitos , Glicoproteínas de Membrana/metabolismo , Ratones , Neoplasias Experimentales/inmunología , Células TH1/inmunología , Células Th2/inmunología , Transfección , Antígeno CD83
8.
PLoS One ; 8(7): e69620, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936061

RESUMEN

Neonatal necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease of preterm infants. Increased intestinal epithelium permeability is an early event in NEC pathogenesis. Autophagy and apoptosis are induced by multiple stress pathways which may impact the intestinal barrier, and they have been associated with pathogenesis of diverse gastrointestinal diseases including inflammatory bowel disease. Using both in vitro and in vivo models, this study investigates autophagy and apoptosis under experimental NEC stresses. Furthermore this study evaluates the effect of erythropoietin (Epo), a component of breast milk previously shown to decrease the incidence of NEC and to preserve intestinal barrier function, on intestinal autophagy and apoptosis. It was found that autophagy and apoptosis are both rapidly up regulated in NEC in vivo as indicated by increased expression of the autophagy markers Beclin 1 and LC3II, and by evidence of apoptosis by TUNEL and cleaved caspase-3 staining. In the rat NEC experimental model, autophagy preceded the onset of apoptosis in intestine. In vitro studies suggested that Epo supplementation significantly decreased both autophagy and apoptosis via the Akt/mTOR signaling pathway and the MAPK/ERK pathway respectively. These results suggest that Epo protects intestinal epithelium from excessive autophagy and apoptosis in experimental NEC.


Asunto(s)
Apoptosis , Autofagia , Enterocolitis Necrotizante/prevención & control , Células Epiteliales/efectos de los fármacos , Eritropoyetina/farmacología , Mucosa Intestinal/efectos de los fármacos , Intestinos/efectos de los fármacos , Animales , Animales Recién Nacidos , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Caspasa 3/genética , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/genética , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Eritropoyetina/genética , Eritropoyetina/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Expresión Génica , Humanos , Recién Nacido , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Intestinos/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Leche Humana/química , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
9.
PLoS One ; 8(5): e65108, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23717690

RESUMEN

Balance among the complex interactions of the gut microbial community is important for intestinal health. Probiotic bacteria can improve bacterial balance and have been used to treat gastrointestinal diseases. Neonatal necrotizing enterocolitis (NEC) is a life-threatening inflammatory bowel disorder primarily affecting premature infants. NEC is associated with extensive inflammatory NF-κB signaling activation as well as intestinal barrier disruption. Clinical studies have shown that probiotic administration may protect against NEC, however there are safety concerns associated with the ingestion of large bacterial loads in preterm infants. Bacteria-free conditioned media (CM) from certain probiotic organisms have been shown to retain bioactivity including anti-inflammatory and cytoprotective properties without the risks of live organisms. We hypothesized that the CM from Lactobacillus acidophilus (La), Bifidobacterium infantis (Bi), and Lactobacillus plantarum (Lp), used separately or together would protect against NEC. A rodent model with intestinal injury similar to NEC was used to study the effect of CM from Lp, La/Bi, and La/Bi/Lp on the pathophysiology of NEC. All the CM suppressed NF-κB activation via preserved IκBα expression and this protected IκBα was associated with decreased liver activity of the proteasome, which is the degrading machinery for IκBα. These CM effects also caused decreases in intestinal production of the pro-inflammatory cytokine TNF-α, a downstream target of the NF-κB pathway. Combined La/Bi and La/Bi/Lp CM in addition protected intestinal barrier function by maintaining tight junction protein ZO-1 levels and localization at the tight junction. Double combined La/Bi CM significantly reduced intestinal injury incidence from 43% to 28% and triple combined La/Bi/Lp CM further reduced intestinal injury incidence to 20%. Thus, this study demonstrates different protective mechanisms and synergistic bioactivity of the CM from different organisms in ameliorating NEC-like intestinal injury in an animal model.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Enterocolitis Necrotizante/prevención & control , Probióticos/administración & dosificación , Animales , Animales Recién Nacidos , Enterocolitis Necrotizante/metabolismo , Femenino , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Intestinos/microbiología , Intestinos/patología , FN-kappa B/metabolismo , Embarazo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ratas , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Proteína de la Zonula Occludens-1/metabolismo
10.
J Biol Chem ; 281(45): 33971-81, 2006 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-16959768

RESUMEN

Metastasis is a primary cause of mortality due to cancer. Early metastatic growth involves both a remodeling of the extracellular matrix surrounding tumors and invasion of tumors across the basement membrane. Up-regulation of extracellular matrix degrading proteases such as urokinase plasminogen activator (uPA) and matrix metalloproteinases has been reported to facilitate tumor cell invasion. Autocrine transforming growth factor-beta (TGF-beta) signaling may play an important role in cancer cell invasion and metastasis; however, the underlying mechanisms remain unclear. In the present study, we report that autocrine TGF-beta supports cancer cell invasion by maintaining uPA levels through protein secretion. Interestingly, treatment of paracrine/exogenous TGF-beta at higher concentrations than autocrine TGF-beta further enhanced uPA expression and cell invasion. The enhanced uPA expression by exogenous TGF-beta is a result of increased uPA mRNA expression due to RNA stabilization. We observed that both autocrine and paracrine TGF-beta-mediated regulation of uPA levels was lost upon depletion of Smad4 protein by RNA interference. Thus, through the Smad pathway, autocrine TGF-beta maintains uPA expression through facilitated protein secretion, thereby supporting tumor cell invasiveness, whereas exogenous TGF-beta further enhances uPA expression through mRNA stabilization leading to even greater invasiveness of the cancer cells.


Asunto(s)
Comunicación Autocrina , Neoplasias de la Mama/patología , Comunicación Paracrina , Estabilidad del ARN , Proteína Smad4/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Northern Blotting , Neoplasias de la Mama/metabolismo , Membrana Celular/metabolismo , Proliferación Celular , Colágeno , Combinación de Medicamentos , Femenino , Genes Dominantes , Humanos , Immunoblotting , Laminina , Metaloproteinasa 9 de la Matriz/metabolismo , Invasividad Neoplásica , Inhibidor 1 de Activador Plasminogénico/metabolismo , Proteoglicanos , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Células Tumorales Cultivadas
11.
J Surg Res ; 117(2): 296-305, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15047135

RESUMEN

BACKGROUND: Smad proteins play a key role in TGF-beta signaling that regulates cell proliferation, differentiation, and apoptosis. Mice deficient in Smad3 develop colonic adenocarcinoma. MATERIALS AND METHODS: We developed a Smad3-deficient colonocyte cell line that was used to study TGF-beta-mediated growth inhibition and induction of apoptosis was compared to young adult mouse colonocyte (YAMC) control cells. Growth inhibition was assessed by cell count and ((3)H)-thymidine incorporation assay. Transcriptional response to TGF-beta was measured by transfecting the reporters p3TP-Lux and p(CAGA)(9)-MLP-luc. TGF-beta-induced apoptosis was assessed using ELISA and Hoechst staining. Mediators of cell-cycle arrest and apoptosis were assayed by Western blot. RESULTS: Smad3-/- cells were resistant to TGF-beta-mediated growth inhibition compared to control cells. Ninety-eight percent of cell count growth inhibition observed in YAMC cells, while 34% inhibition was observed in Smad3-/- cells after TGF-beta treatment. ((3)H)-thymidine incorporation was inhibited by 61% in YAMC cells, while Smad3-/- cells showed 25% inhibition after TGF-beta treatment. Smad3-/- cells were deficient in luciferase reporter induction by TGF-beta. TGF-beta induced apoptosis 8-fold in YAMC cells, but had no effect on apoptosis in Smad3-/- cells. p21(Cip11) and PAI-1 are induced in YAMC cells by TGF-beta, but unchanged in Smad3-/- cells. TGF-beta decreases cyclin D1 levels in YAMC cells but does not affect levels in Smad3-/- cells. CONCLUSIONS: Our findings suggest that the loss of Smad3 contributes to resistance of TGF-beta growth inhibition and apoptosis in colonic epithelium. This may represent a mechanism by which cells are able to escape antiproliferative controls and embark on a pathway toward neoplasia.


Asunto(s)
Apoptosis/fisiología , Colon/fisiología , Proteínas de Unión al ADN/fisiología , Inhibidores de Crecimiento/fisiología , Mucosa Intestinal/fisiología , Transactivadores/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , División Celular/fisiología , Línea Celular , Proteínas de Unión al ADN/deficiencia , Ratones , Ratones Transgénicos , Mutación , Regiones Promotoras Genéticas/fisiología , Proteína smad3 , Transactivadores/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA