Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Dev Biol ; 338(2): 270-9, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20025862

RESUMEN

Anterior Gradient 2 (AGR2) is a protein disulfide isomerase that plays important roles in diverse processes in multiple cell lineages as a developmental regulator, survival factor and susceptibility gene for inflammatory bowel disease. Here, we show using germline and inducible Agr2-/- mice that Agr2 plays important roles in intestinal homeostasis. Agr2-/- intestine has decreased goblet cell Mucin 2, dramatic expansion of the Paneth cell compartment, abnormal Paneth cell localization, elevated endoplasmic reticulum (ER) stress, severe terminal ileitis and colitis. Cell culture experiments show that Agr2 expression is induced by ER stress, and that siRNA knockdown of Agr2 increases ER stress response. These studies implicate Agr2 in intestinal homeostasis and ER stress and suggest a role in the etiology of inflammatory bowel disease.


Asunto(s)
Retículo Endoplásmico/patología , Células Caliciformes/patología , Homeostasis , Mucoproteínas/genética , Células de Paneth/patología , Animales , Retículo Endoplásmico/metabolismo , Enfermedades Inflamatorias del Intestino/etiología , Intestinos/química , Intestinos/patología , Ratones , Ratones Noqueados , Mucina 2/análisis , Mucoproteínas/deficiencia , Proteínas Oncogénicas , Estrés Fisiológico
2.
Cancer Res ; 65(19): 8662-70, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16204034

RESUMEN

Germ line DNA mismatch repair mutations in MLH1 and MSH2 underlie the vast majority of hereditary non-polyposis colon cancer. Four mammalian homologues of Escherichia coli MutL heterodimerize to form three distinct complexes: MLH1/PMS2, MLH1/MLH3, and MLH1/PMS1. Although MLH1/PMS2 is generally thought to have the major MutL activity, the precise contributions of each MutL heterodimer to mismatch repair functions are poorly understood. Here, we show that Mlh3 contributes to mechanisms of tumor suppression in the mouse. Mlh3 deficiency alone causes microsatellite instability, impaired DNA-damage response, and increased gastrointestinal tumor susceptibility. Furthermore, Mlh3;Pms2 double-deficient mice have tumor susceptibility, shorter life span, microsatellite instability, and DNA-damage response phenotypes that are indistinguishable from Mlh1-deficient mice. Our data support previous results from budding yeast that show partial functional redundancy between MLH3 and PMS2 orthologues for mutation avoidance and show a role for Mlh3 in gastrointestinal and extragastrointestinal tumor suppression. The data also suggest a mechanistic basis for the more severe mismatch repair-related phenotypes and cancer susceptibility in Mlh1- versus Mlh3- or Pms2-deficient mice. Contributions by both MLH1/MLH3 and MLH1/PMS2 complexes to mechanisms of mismatch repair-mediated tumor suppression, therefore, provide an explanation why, among MutL homologues, only germ line mutations in MLH1 are common in hereditary non-polyposis colon cancer.


Asunto(s)
Adenosina Trifosfatasas/genética , Disparidad de Par Base , Proteínas Portadoras/genética , Enzimas Reparadoras del ADN/genética , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Neoplasias Gastrointestinales/genética , Proteínas Adaptadoras Transductoras de Señales , Adenosina Trifosfatasas/deficiencia , Animales , Proteínas Portadoras/biosíntesis , Enzimas Reparadoras del ADN/deficiencia , Proteínas de Unión al ADN/deficiencia , Neoplasias Gastrointestinales/patología , Genes Supresores de Tumor , Predisposición Genética a la Enfermedad , Ratones , Ratones Endogámicos C57BL , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto , Homólogo 1 de la Proteína MutL , Proteínas MutL , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética
3.
Cancer Discov ; 2(6): 540-53, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22628407

RESUMEN

Colorectal cancer is a classic example of a tumor that progresses through multiple distinct stages in its evolution. To understand the mechanisms regulating the transition from indolent to invasive disease, we profiled somatic copy number alterations in noninvasive adenomas and invasive adenocarcinomas from Apc and DNA mismatch repair (MMR) mutant mouse models. We identified a recurrent amplicon on mouse chromosome 8 that encodes microRNA (miRNA) 23a and -27a (miR). miR-23a and -27a levels are upregulated in mouse intestinal adenocarcinomas, primary tumors from patients with stage I/II colorectal cancers, as well as in human colorectal cancer cell lines and cancer stem cells. Functionally, miR-23a promotes the migration and invasion of colorectal cancer cells and stem cells, whereas miR-27a primarily promotes proliferation. We computationally and experimentally validated that metastasis suppressor 1 (MTSS1) is a direct miR-23a target and similarly validated that the ubiquitin ligase FBXW7 is a direct miR-27a target. Analyses of computationally predicted target genes in microarray data sets of patients with colorectal cancers are consistent with a role for miR-23a, but not miR-27a, specifically in invasive colorectal cancers.


Asunto(s)
Adenocarcinoma/genética , Adenoma/genética , Neoplasias Colorrectales/genética , MicroARNs/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenoma/metabolismo , Adenoma/patología , Animales , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Variaciones en el Número de Copia de ADN , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Invasividad Neoplásica/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
4.
Oncotarget ; 1(7): 596-605, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21317455

RESUMEN

Colorectal cancer metastatic recurrence and chemoresistance are major causes of morbidity and mortality. Colon cancer initiating cells (CCIC) are thought to contribute to both these processes. To identify drugs with anti-CCIC activity we screened a number of FDA approved and investigational compounds. We found that the class I selective histone deacetylase inhibitor (HDACi) MGCD0103 has significant activity against CCIC, and also significantly inhibits non-CCIC CRC cell xenograft formation. Both MGCD0103 and the pan-HDAC inhibitor Trichostatin impairs CCIC clonogenicity and cause cell cycle arrest and cell death. Gene expression profiling revealed that the canonical WNT ligand DKK-1 is a highly upregulated target of HDAC inhibitors. Despite the presence of APC mutations and constitutive WNT signaling in CCIC, both transfected and recombinant DKK-1 dramatically inhibit CCIC proliferation and clonogenicity. Overall, these data show that inhibition of class I HDACs is a promising novel approach to target both CCIC and non-CCIC CRC cells. Our studies also provide novel insights into roles for DKK1 in addition to canonical WNT signaling and the mechanism of CCIC tumor formation.


Asunto(s)
Apoptosis/efectos de los fármacos , Benzamidas/farmacología , Carcinoma/patología , Ciclo Celular/efectos de los fármacos , Neoplasias del Colon/patología , Péptidos y Proteínas de Señalización Intercelular/genética , Células Madre Neoplásicas/efectos de los fármacos , Pirimidinas/farmacología , Proteínas Wnt/genética , Animales , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Cultivadas , Evaluación Preclínica de Medicamentos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HT29 , Células HeLa , Inhibidores de Histona Desacetilasas/clasificación , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Células Madre Neoplásicas/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Proteínas Wnt/metabolismo
5.
Cancer Res ; 70(4): 1469-78, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20145124

RESUMEN

NOTCH signaling is critical for specifying the intestinal epithelial cell lineage and for initiating colorectal adenomas and colorectal cancers (CRC). Based on evidence that NOTCH is important for the maintenance and self-renewal of cancer-initiating cells in other malignancies, we studied the role of NOTCH signaling in colon cancer-initiating cells (CCIC). Tumors formed by CCICs maintain many properties of the primary CRCs from which they were derived, such as glandular organization, cell polarity, gap junctions, and expression of characteristic CRC molecular markers. Furthermore, CCICs have the property of self-renewal. In this study, we show that NOTCH signaling is 10- to 30-fold higher in CCIC compared with widely used colon cancer cell lines. Using small-molecule inhibition and short hairpin RNA knockdown, we show that NOTCH prevents CCIC apoptosis through repression of cell cycle kinase inhibitor p27 and transcription factor ATOH1. NOTCH is also critical to intrinsic maintenance of CCIC self-renewal and the repression of secretory cell lineage differentiation genes such as MUC2. Our findings describe a novel human cell system to study NOTCH signaling in CRC tumor initiation and suggest that inhibition of NOTCH signaling may improve CRC chemoprevention and chemotherapy.


Asunto(s)
Carcinoma/genética , Diferenciación Celular/genética , Proliferación Celular , Transformación Celular Neoplásica/genética , Neoplasias del Colon/genética , Células Enteroendocrinas/fisiología , Células Madre Neoplásicas/fisiología , Receptor Notch1/fisiología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/fisiología , Carcinoma/patología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Neoplasias del Colon/patología , Células Enteroendocrinas/efectos de los fármacos , Células Enteroendocrinas/metabolismo , Células Enteroendocrinas/patología , Células HCT116 , Células HT29 , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , ARN Interferente Pequeño/farmacología , Receptor Notch1/antagonistas & inhibidores , Receptor Notch1/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cancer Prev Res (Phila) ; 3(5): 597-603, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20403997

RESUMEN

Genetic variation in 3-hydroxy-3-methylglutaryl CoA reductase (HMGCR), the rate-limiting enzyme in cholesterol synthesis, modifies the effect of statins on serum cholesterol levels. Long-term use of statins is associated with a reduced risk of colorectal cancer (CRC) in some, but not all, studies. We genotyped variants in 40 candidate genes important for cholesterol synthesis and metabolism in a population-based case-control study of CRC involving 2,138 incident cases and 2,049 population-based controls. We identified a single-nucleotide polymorphism in the HMGCR gene that significantly modified the protective association between statins and CRC risk. Compared with nonusers, the unadjusted odds ratio of CRC among statin users with the A/A genotype of rs12654264 in HMGCR was 0.3 (95% confidence interval, 0.18-0.51) and among statin users with the T/T genotype was 0.66 (95% confidence interval, 0.41-1.06; P-interaction = 0.0012). This genetic variant (A/A genotype of rs12654264) also was associated with lower serum levels of low-density lipoprotein among all cases and controls. In colon cancer cell lines, the reduction in cholesterol levels after statin treatment was substantially stronger in cells carrying the A/A genotype, and this difference was related to alternative splicing involving the HMGCR statin-binding domain. We anticipate that these data may advance the development of personalized statin use for reducing the risk of cancer as well as cardiovascular disease among the approximately 25 million people currently using statins worldwide.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Resistencia a Antineoplásicos/genética , Hidroximetilglutaril-CoA Reductasas/genética , Anciano , Empalme Alternativo , Anticolesterolemiantes/uso terapéutico , Estudios de Casos y Controles , LDL-Colesterol/sangre , Neoplasias Colorrectales/enzimología , Femenino , Genotipo , Humanos , Hidroximetilglutaril-CoA Reductasas/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Masculino , Polimorfismo de Nucleótido Simple
7.
Dev Dyn ; 237(2): 316-27, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18163532

RESUMEN

E. coli AlkB has been intensively studied since 1983, but the in vivo roles of its mammalian homologue Alkbh1 are unknown. We, therefore, created null mice for Alkbh1. Alkbh1 mRNA is expressed at highest levels in the trophoblast lineages of the developing placenta. Alkbh1(-/-) placentas have decreased expression of differentiated trophoblast markers including Tpbp, Gcm1, and Pl-1, and increased expression of the trophoblast stem cell marker Eomes. Alkbh1 localizes to nuclear euchromatin, and interacts strongly with Mrj, an essential placental gene that mediates gene repression by recruitment of class II histone deacetylases (HDACs). Competition experiments show Alkbh1 and HDAC4 binding to Mrj are mutually exclusive, which causes decreased HDAC activity and increased target gene expression. Our study demonstrates Alkbh1 performs important functions in placental trophoblast lineage differentiation and participates in mechanisms of transcriptional regulation.


Asunto(s)
Diferenciación Celular/fisiología , Dioxigenasas/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Filogenia , Placenta/citología , Factores de Transcripción/genética , Trofoblastos/fisiología , Histona H2a Dioxigenasa, Homólogo 1 de AlkB , Animales , Diferenciación Celular/genética , Análisis por Conglomerados , ADN-(Sitio Apurínico o Apirimidínico) Liasa , Eucromatina/metabolismo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Proteínas del Choque Térmico HSP40/metabolismo , Inmunoprecipitación , Hibridación in Situ , Ratones , Ratones Noqueados , Modelos Genéticos , Chaperonas Moleculares/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA