Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Biochim Biophys Acta Mol Cell Res ; 1864(12): 2347-2355, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28893557

RESUMEN

Intestinal epithelial cells form a protective barrier in limiting gut luminal content potentially harmful to the host. Upon gut epithelium injury, several signals instruct epithelial cells to undergo a rapid healing process. Defects in this process induce inflammatory responses and can further evolve into chronic gut inflammatory diseases. We previously identified the transcription factor CUX1 as crucial for protecting against experimental colitis in mice. However, the precise molecular mechanisms by which CUX1 intervenes during this biological process are unknown. Our aim was to evaluate CUX1 biological and functional roles during intestinal epithelial cell wound healing. RNAi knockdown of CUX1 in intestinal epithelial cells revealed a crucial role for this regulator in migratory response following wounding assays. Gene expression profiling identified several gene transcripts modulated in absence of CUX1 during wound healing for which a significant number was associated with cell motility and cytoskeleton function. Chromatin immunoprecipitation assays identified the guanine nucleotide exchange factor Vav2 gene as a direct target for CUX1. Coincidently, reduction of VAV2 in absence of CUX1 was associated with a significant decrease of RAC1 activity in response to epithelial wounding. Our results identify a novel pathway by which CUX1 regulates normal intestinal epithelial cell restitution.


Asunto(s)
Proteínas de Homeodominio/genética , Inflamación/genética , Neuropéptidos/genética , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Represoras/genética , Cicatrización de Heridas/genética , Proteína de Unión al GTP rac1/genética , Animales , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Humanos , Inflamación/patología , Mucosa Intestinal/metabolismo , Intestinos/patología , Ratones
2.
Am J Physiol Gastrointest Liver Physiol ; 300(4): G586-97, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21212325

RESUMEN

The regulation of intestinal epithelial cell adhesion and migratory properties is often compromised in inflammatory bowel disease (IBD). Despite an increasing interest in bone morphogenetic protein (Bmp) signaling in gut pathologies, little is known of the specific roles played by individual Smads in intestinal epithelial functions. In the present study, we generated a mouse model with deletion of Smad5 transcriptional effector of the Bmp signaling pathway exclusively in the intestinal epithelium. Proliferation, migration, and apical junctional complex (AJC) protein expression were analyzed by immunofluorescence and Western blot. Human intestinal biopsies from control and IBD patients were analyzed for SMAD5 gene transcript expression by quantitative PCR (qPCR). Smad5(ΔIEC) and control mice were subjected to dextran sulfate sodium (DSS)-induced experimental colitis, and their clinical and histological symptoms were assessed. Loss of Smad5 led to intestinal epithelial hypermigration and deregulation of the expression of claudin-1 and claudin-2. E-cadherin was found to be equally expressed but displaced from the AJC to the cytoplasm in Smad5(ΔIEC) mice. Analysis of SMAD5 gene expression in human IBD patient samples revealed a significant downregulation of the gene transcript in Crohn's disease and ulcerative colitis samples. Smad5(ΔIEC) mice exposed to experimental DSS colitis were significantly more susceptible to the disease and had impaired wound healing during the recovery phase. Our results support that Smad5 is partly responsible for mediating Bmp signals in intestinal epithelial cells. In addition, deficiency in epithelial Smad5 leads to the deregulation of cell migration by disassembling the AJC with increasing susceptibility to experimental colitis and impairment in wound healing.


Asunto(s)
Colitis/metabolismo , Susceptibilidad a Enfermedades/metabolismo , Uniones Intercelulares/metabolismo , Mucosa Intestinal/metabolismo , Proteína Smad5/metabolismo , Animales , Western Blotting , Movimiento Celular/genética , Colitis/inducido químicamente , Colitis/genética , Colitis/patología , Susceptibilidad a Enfermedades/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Técnica del Anticuerpo Fluorescente , Humanos , Uniones Intercelulares/genética , Uniones Intercelulares/patología , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Proteína Smad5/genética
3.
J Biol Chem ; 284(37): 25220-9, 2009 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-19608741

RESUMEN

Stem cells of the gut epithelium constantly produce precursors that progressively undergo a succession of molecular changes resulting in growth arrest and commitment to a specific differentiation program. Few transcriptional repressors have been identified that maintain the normal intestinal epithelial cell (IEC) proliferation state. Herein, we show that the nuclear receptor co-repressor (NCoR1) is differentially expressed during the proliferation-to-differentiation IEC transition. Silencing of NCoR1 expression in proliferating cells of crypt origin resulted in a rapid growth arrest without associated cell death. A genechip profiling analysis identified several candidate genes to be up-regulated in NCoR1-deficient IEC. Pigment epithelium-derived factor (PEDF, also known as serpinf1), a suspected tumor suppressor gene that plays a key role in the inhibition of epithelial tissue growth, was significantly up-regulated in these cells. Chromatin immunoprecipitation experiments showed that the PEDF gene promoter was occupied by NCoR1 in proliferating epithelial cells. Multiple retinoid X receptor (RXR) heterodimers interacting sites of the PEDF promoter were confirmed to interact with RXR and retinoid acid receptor (RAR). Cotransfection assays showed that RXR and RAR were able to transactivate the PEDF promoter and that NCoR1 was repressing this effect. Finally, forced expression of PEDF in IEC resulted in a slower rate of proliferation. These observations suggest that NCoR1 expression is required to maintain IEC in a proliferative state and identify PEDF as a novel transcriptional target for NCoR1 repressive action.


Asunto(s)
Células Epiteliales/metabolismo , Proteínas del Ojo/metabolismo , Regulación de la Expresión Génica , Intestinos/citología , Factores de Crecimiento Nervioso/metabolismo , Proteínas Nucleares/fisiología , Proteínas Represoras/fisiología , Serpinas/metabolismo , Secuencia de Aminoácidos , Animales , Células CACO-2 , Proliferación Celular , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Co-Represor 1 de Receptor Nuclear , Ratas , Fracciones Subcelulares/metabolismo
4.
Am J Physiol Gastrointest Liver Physiol ; 297(1): G124-34, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19389805

RESUMEN

Hepatocyte nuclear factor 4alpha (HNF4alpha) is a regulator of hepatocyte and pancreatic transcription. Hnf4alpha deletion in the mouse is embryonically lethal with severe defects in visceral endoderm formation. It has been concluded in the past that the role of Hnf4alpha in the developing colon was much less important than in the liver. However, the precise role of Hnf4alpha in the homeostasis of the small intestinal epithelium remains unclear. Our aim was to evaluate the potential of Hnf4alpha to support an intestinal epithelial phenotype. First, Hnf4alpha potential to dictate this phenotype was assessed in nonintestinal cell lines in vitro. Forced expression of Hnf4alpha in fibroblasts showed an induction of features normally restricted to epithelial cells. Combinatory expression of Hnf4alpha with specific transcriptional regulators of the intestine resulted in the induction of intestinal epithelial genes in this context. Second, the importance of Hnf4alpha in maintaining the homeostasis of the intestinal epithelium was investigated in mice. Mice conditionally deficient for intestinal Hnf4alpha developed normally throughout adulthood with an epithelium displaying normal morphological and functional structures with minor alterations. Subtle but statistical differences were observed at the proliferation and the cytodifferentiation levels. Hnf4alpha mutant mice displayed an increase in the number of goblet and enteroendocrine cells compared with controls. Given the fundamental role of this transcription factor in other tissues, these findings dispute the crucial role for this regulator in the maintenance of intestinal epithelial cell function at a period of time that follows cytodifferentiation but may suggest a functional role in instructing cells to become specific to the intestinal epithelium.


Asunto(s)
Diferenciación Celular , Células Epiteliales/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Mucosa Intestinal/metabolismo , Envejecimiento/metabolismo , Animales , Factor de Transcripción CDX2 , Células CACO-2 , Proliferación Celular , Forma de la Célula , Células Epiteliales/diagnóstico por imagen , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Factor Nuclear 4 del Hepatocito/deficiencia , Factor Nuclear 4 del Hepatocito/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Homeostasis , Humanos , Integrasas/genética , Mucosa Intestinal/ultraestructura , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Células 3T3 NIH , Fenotipo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección , Ultrasonografía
5.
Sci Rep ; 9(1): 4200, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30862908

RESUMEN

Mutations in the HNF4A gene cause MODY1 and are associated with an increased risk of Type 2 diabetes mellitus. On the other hand, incretins are hormones that potentiate reductions in blood glucose levels. Given the established role of incretin-based therapy to treat diabetes and metabolic disorders, we investigated a possible regulatory link between intestinal epithelial HNF4α and glucose-dependent insulinotropic polypeptide (GIP), an incretin that is specifically produced by gut enteroendocrine cells. Conditional deletion of HNF4α in the whole intestinal epithelium was achieved by crossing Villin-Cre and Hnf4αloxP/loxP C57BL/6 mouse models. GIP expression was measured by qPCR, immunofluorescence and ELISA. Gene transcription was assessed by luciferase and electrophoretic mobility shift assays. Metabolic parameters were analyzed by indirect calorimetry and dual-energy X-ray absorptiometry. HNF4α specific deletion in the intestine led to a reduction in GIP. HNF4α was able to positively control Gip transcriptional activity in collaboration with GATA-4 transcription factor. Glucose homeostasis and glucose-stimulated insulin secretion remained unchanged in HNF4α deficient mice. Changes in GIP production in these mice did not impact nutrition or energy metabolism under normal physiology but led to a reduction of bone area and mineral content, a well described physiological consequence of GIP deficiency. Our findings point to a novel regulatory role between intestinal HNF4α and GIP with possible functional impact on bone density.


Asunto(s)
Células Enteroendocrinas/metabolismo , Polipéptido Inhibidor Gástrico/biosíntesis , Factor Nuclear 4 del Hepatocito/metabolismo , Mucosa Intestinal/metabolismo , Transcripción Genética , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Polipéptido Inhibidor Gástrico/genética , Eliminación de Gen , Factor Nuclear 4 del Hepatocito/genética , Ratones , Ratones Transgénicos
6.
FASEB J ; 21(14): 3853-65, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17622569

RESUMEN

Intestinal epithelial integrity and polarity are maintained by cohesive interactions between cells via the formation of tight junctions. Irregularities in tight junctions have only recently been found to be associated with the initiation and progression of intestinal neoplasia. The claudin family of proteins is integral to the structure and function of the tight junction but little is known of the molecular events that regulate the expression of these components. The present report identifies cathepsin L, classically a lysosomal cysteine protease, as being induced during intestinal epithelial cell polarization and differentiation. Inhibition of intracellular cathepsin L activity results in the accumulation of disorganized cell layers and a decline in the expression of differentiation markers in cultured intestinal epithelial cells. This coincides with a rapid up-regulation of claudin-1 protein accumulation. Mutant mice defective in cathepsin L activity (furless) display an elevated level of intestinal claudin-1 and claudin-2 expression. Loss of cathepsin L activity leads to a marked increase in tumor multiplicity in the intestine of Apc(Min) mice. Given the traditionally viewed biological role of cathepsin L in the processing of lysosomal content as well as in pathological extracellular matrix remodeling, the results here demonstrate an as yet unsuspected intracellular role for this protease in normal intestinal epithelial polarization and initiation of neoplasia.


Asunto(s)
Catepsinas/antagonistas & inhibidores , Catepsinas/metabolismo , Cisteína Endopeptidasas/metabolismo , Neoplasias Intestinales/etiología , Neoplasias Intestinales/genética , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Animales , Secuencia de Bases , Células CACO-2 , Catepsina L , Catepsinas/deficiencia , Catepsinas/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular , Claudina-1 , Cisteína Endopeptidasas/deficiencia , Cisteína Endopeptidasas/genética , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Predisposición Genética a la Enfermedad , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/enzimología , Neoplasias Intestinales/metabolismo , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Datos de Secuencia Molecular , Inhibidores de Proteasas/farmacología , Conejos , Regulación hacia Arriba/fisiología
7.
Inflamm Bowel Dis ; 16(10): 1739-50, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20848487

RESUMEN

BACKGROUND: Cux1 is a ubiquitous transcriptional factor that has been associated with cell proliferation, migration, invasion, and differentiation. Cux1 is an effector of the transforming growth factor beta (TGFß) pathway, PAR(2) receptor signaling, and cellular migration, mechanisms intimately related to inflammatory bowel diseases (IBD). METHODS: CD1 mice treated with dextran sulfate sodium (DSS) in drinking water and cultured intestinal epithelial cells were used to determine Cux1 expression under inflammatory conditions. A commercial cDNA library was used to monitor CUX1 expression in IBD patients. The Cux1(ΔHD/ΔHD) hypomorphic mouse model (Cux1ΔHD) treated with DSS in drinking water was used and the disease severity assessed. RESULTS: Cux1 expression increased in cultured intestinal epithelial cells stimulated with tumor necrosis factor alpha (TNFα), in the mouse intestinal epithelium during experimental colitis and in human IBD patient samples. DSS-induced colitis in Cux1ΔHD mice was more severe according to clinical observations such as weight loss, colon length, and rectal bleeding. Histological observations confirmed an increase of IBD-related morphological changes including ulceration and mucosal infiltration of leukocytes in Cux1ΔHD mice. An increased number of pSer(276)-RelA-positive cells and higher expression levels of proinflammatory cytokines were also measured in the colon of Cux1ΔHD diseased animals. Elevated levels of Cxcl1 were measured before and after DSS-treatment and a greater neutrophilic infiltration was quantified in DSS-treated Cux1ΔHD mice. Finally, mucosal healing was significantly impaired in Cux1ΔHD mice during recovery from DSS treatment. CONCLUSIONS: CUX1 is increased in response to inflammatory stress and its nuclear expression is crucial to protect against DSS-induced colitis and subsequent mucosal healing.


Asunto(s)
Colitis/prevención & control , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/fisiología , Proteínas de Homeodominio/genética , Enfermedades Inflamatorias del Intestino/genética , Mucosa Intestinal/metabolismo , Proteínas Nucleares/genética , Proteínas Represoras/genética , Animales , Colitis/inducido químicamente , Colitis/patología , Sulfato de Dextran/toxicidad , Técnica del Anticuerpo Fluorescente , Biblioteca de Genes , Proteínas de Homeodominio/metabolismo , Humanos , Immunoblotting , Técnicas para Inmunoenzimas , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Membrana Mucosa/efectos de los fármacos , Membrana Mucosa/metabolismo , Membrana Mucosa/patología , Proteínas Nucleares/metabolismo , ARN Mensajero/genética , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción , Factor de Necrosis Tumoral alfa/farmacología
8.
FEBS J ; 277(20): 4241-53, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20945532

RESUMEN

The CCAAT-Displacement-Protein (CUX1) can transcriptionally repress sucrase­isomaltase gene expression, a specific product of enterocytes that becomes re-expressed during human colonic polyposis. Little is known of the gene repertoire that is directly affected by CUX1 in the intestinal epithelial context. This article identifies the Promyelocytic Leukemia Zinc Finger (PLZF) gene as a transcriptional target for the CUX1 repressor. CUX1 interacts in vivo with multiple DNA-binding sites in the 5'-UTR and promoter of the PLZF gene in colorectal cancer cells, a region that is functionally targeted by CUX1 in cotransfection assays. PLZF was found to be induced in colorectal cancer cell lines, correlating with a low detectable level of CUX1, a pattern that was reversed in normal human colonocytes. Reduction of p200CUX1 expression by RNAi in the Caco-2/15 cell line increased PLZF gene transcript expression. Because of the implication of Plzf in the regulation of stem cell maintenance, as well as Wnt and Ras signaling, in other systems, our observations suggest that the novel genetic relationship between CUX1 and PLZF could be of relevance to human diseases, such as leukemia, and open up a new field of investigation for the implication of these regulators during intestinal polyposis and cancer.


Asunto(s)
Neoplasias Colorrectales/genética , Proteínas de Homeodominio/genética , Factores de Transcripción de Tipo Kruppel/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Transcripción Genética , Regiones no Traducidas 5' , Sitios de Unión , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Sistemas de Liberación de Medicamentos , Humanos , Regiones Promotoras Genéticas , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Factores de Transcripción , Dedos de Zinc/genética
9.
Cancer Res ; 70(22): 9423-33, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21062980

RESUMEN

Hepatocyte nuclear factor-4α (Hnf4α) is a transcription factor that controls epithelial cell polarity and morphogenesis. Hnf4α conditional deletion during postnatal development has minor effects on intestinal epithelium integrity but promotes activation of the Wnt/ß-catenin pathway without causing tumorigenesis. Here, we show that Hnf4α does not act as a tumor-suppressor gene but is crucial in promoting gut tumorigenesis in mice. Polyp multiplicity in ApcMin mice lacking Hnf4α is suppressed compared with littermate ApcMin controls. Analysis of microarray gene expression profiles from mice lacking Hnf4α in the intestinal epithelium identifies novel functions of this transcription factor in targeting oxidoreductase-related genes involved in the regulation of reactive oxygen species (ROS) levels. This role is supported with the demonstration that HNF4α is functionally involved in the protection against spontaneous and 5-fluorouracil chemotherapy-induced production of ROS in colorectal cancer cell lines. Analysis of a colorectal cancer patient cohort establishes that HNF4α is significantly upregulated compared with adjacent normal epithelial resections. Several genes involved in ROS neutralization are also induced in correlation with HNF4A expression. Altogether, the findings point to the nuclear receptor HNF4α as a potential therapeutic target to eradicate aberrant epithelial cell resistance to ROS production during intestinal tumorigenesis.


Asunto(s)
Perfilación de la Expresión Génica , Factor Nuclear 4 del Hepatocito/metabolismo , Mucosa Intestinal/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Adenoma/genética , Adenoma/metabolismo , Adenoma/patología , Animales , Antimetabolitos Antineoplásicos/farmacología , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Femenino , Fluorouracilo/farmacología , Células HCT116 , Células HT29 , Factor Nuclear 4 del Hepatocito/genética , Humanos , Immunoblotting , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Neoplasias Intestinales/genética , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Pólipos Intestinales/genética , Pólipos Intestinales/metabolismo , Pólipos Intestinales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
PLoS One ; 4(10): e7609, 2009 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-19898610

RESUMEN

BACKGROUND: Hnf4alpha, an epithelial specific transcriptional regulator, is decreased in inflammatory bowel disease and protects against chemically-induced colitis in mice. However, the precise role of this factor in maintaining normal inflammatory homeostasis of the intestine remains unclear. The aim of this study was to evaluate the sole role of epithelial Hnf4alpha in the maintenance of gut inflammatory homeostasis in mice. METHODOLOGY/PRINCIPAL FINDINGS: We show here that specific epithelial deletion of Hnf4alpha in mice causes spontaneous chronic intestinal inflammation leading to focal areas of crypt dropout, increased cytokines and chemokines secretion, immune cell infiltrates and crypt hyperplasia. A gene profiling analysis in diseased Hnf4alpha null colon confirms profound genetic changes in cell death and proliferative behaviour related to cancer. Among the genes involved in the immune protection through epithelial barrier function, we identify the ion transporter claudin-15 to be down-modulated early in the colon of Hnf4alpha mutants. This coincides with a significant decrease of mucosal ion transport but not of barrier permeability in young animals prior to the manifestation of the disease. We confirm that claudin-15 is a direct Hnf4alpha gene target in the intestinal epithelial context and is down-modulated in mouse experimental colitis and inflammatory bowel disease. CONCLUSION: Our results highlight the critical role of Hnf4alpha to maintain intestinal inflammatory homeostasis during mouse adult life and uncover a novel function for Hnf4alpha in the regulation of claudin-15 expression. This establishes Hnf4alpha as a mediator of ion epithelial transport, an important process for the maintenance of gut inflammatory homeostasis.


Asunto(s)
Colitis/inducido químicamente , Colon/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Transporte Iónico , Animales , Claudinas , Eliminación de Gen , Homeostasis , Inflamación , Mucosa Intestinal/metabolismo , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA