Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Oncogene ; 38(25): 5107, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31068667

RESUMEN

In the original version of this article the authors noted that the GEO accession number for the relevant dataset was listed incorrectly as GSE12454.

2.
Oncogene ; 38(25): 5091-5106, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30858547

RESUMEN

The myeloid translocation gene family member MTG16 is a transcriptional corepressor that relies on the DNA-binding ability of other proteins to determine specificity. One such protein is the ZBTB family member Kaiso, and the MTG16:Kaiso interaction is necessary for repression of Kaiso target genes, such as matrix metalloproteinase-7. Using the azoxymethane and dextran sodium sulfate (AOM/DSS) murine model of colitis-associated carcinoma, we previously determined that MTG16 loss accelerates tumorigenesis and inflammation. However, it was unknown whether this effect was modified by Kaiso-dependent transcriptional repression. To test for a genetic interaction between MTG16 and Kaiso in inflammatory carcinogenesis, we subjected single and double knockout (DKO) mice to the AOM/DSS protocol. Mtg16-/- mice demonstrated increased colitis and tumor burden; in contrast, disease severity in Kaiso-/- mice was equivalent to wild-type controls. Surprisingly, Kaiso deficiency in the context of MTG16 loss reversed injury and pro-tumorigenic responses in the intestinal epithelium following AOM/DSS treatment, and tumor numbers were returned to near to wild-type levels. Transcriptomic analysis of non-tumor colon tissue demonstrated that changes induced by MTG16 loss were widely mitigated by concurrent Kaiso loss, and DKO mice demonstrated downregulation of metabolism and cytokine-associated gene sets with concurrent activation of DNA damage checkpoint pathways as compared with Mtg16-/-. Further, Kaiso knockdown in intestinal enteroids reduced stem- and WNT-associated phenotypes, thus abrogating the induction of these pathways observed in Mtg16-/- samples. Together, these data suggest that Kaiso modifies MTG16-driven inflammation and tumorigenesis and suggests that Kaiso deregulation contributes to MTG16-dependent colitis and CAC phenotypes.


Asunto(s)
Adenocarcinoma/genética , Carcinogénesis/genética , Colitis/complicaciones , Colitis/genética , Neoplasias del Colon/genética , Proteínas Represoras/genética , Factores de Transcripción/fisiología , Adenocarcinoma/patología , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Colitis/patología , Neoplasias del Colon/patología , Femenino , Células HCT116 , Células HEK293 , Humanos , Inflamación/complicaciones , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción/genética
3.
J Clin Invest ; 125(7): 2646-60, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26053663

RESUMEN

Patients with inflammatory bowel disease are at increased risk for colon cancer due to augmented oxidative stress. These patients also have compromised antioxidant defenses as the result of nutritional deficiencies. The micronutrient selenium is essential for selenoprotein production and is transported from the liver to target tissues via selenoprotein P (SEPP1). Target tissues also produce SEPP1, which is thought to possess an endogenous antioxidant function. Here, we have shown that mice with Sepp1 haploinsufficiency or mutations that disrupt either the selenium transport or the enzymatic domain of SEPP1 exhibit increased colitis-associated carcinogenesis as the result of increased genomic instability and promotion of a protumorigenic microenvironment. Reduced SEPP1 function markedly increased M2-polarized macrophages, indicating a role for SEPP1 in macrophage polarization and immune function. Furthermore, compared with partial loss, complete loss of SEPP1 substantially reduced tumor burden, in part due to increased apoptosis. Using intestinal organoid cultures, we found that, compared with those from WT animals, Sepp1-null cultures display increased stem cell characteristics that are coupled with increased ROS production, DNA damage, proliferation, decreased cell survival, and modulation of WNT signaling in response to H2O2-mediated oxidative stress. Together, these data demonstrate that SEPP1 influences inflammatory tumorigenesis by affecting genomic stability, the inflammatory microenvironment, and epithelial stem cell functions.


Asunto(s)
Colitis/complicaciones , Neoplasias del Colon/etiología , Selenoproteína P/fisiología , Animales , Antioxidantes/metabolismo , Apoptosis , Neoplasias del Colon/patología , Neoplasias del Colon/fisiopatología , Daño del ADN , Inestabilidad Genómica , Haploinsuficiencia , Macrófagos/clasificación , Macrófagos/patología , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/fisiología , Estrés Oxidativo , Estructura Terciaria de Proteína , Selenio/administración & dosificación , Selenio/metabolismo , Selenoproteína P/deficiencia , Selenoproteína P/genética , Microambiente Tumoral , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/fisiología
4.
PLoS One ; 8(7): e67845, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23861820

RESUMEN

Selenium (Se) is an essential micronutrient that exerts its functions via selenoproteins. Little is known about the role of Se in inflammatory bowel disease (IBD). Epidemiological studies have inversely correlated nutritional Se status with IBD severity and colon cancer risk. Moreover, molecular studies have revealed that Se deficiency activates WNT signaling, a pathway essential to intestinal stem cell programs and pivotal to injury recovery processes in IBD that is also activated in inflammatory neoplastic transformation. In order to better understand the role of Se in epithelial injury and tumorigenesis resulting from inflammatory stimuli, we examined colonic phenotypes in Se-deficient or -sufficient mice in response to dextran sodium sulfate (DSS)-induced colitis, and azoxymethane (AOM) followed by cyclical administration of DSS, respectively. In response to DSS alone, Se-deficient mice demonstrated increased morbidity, weight loss, stool scores, and colonic injury with a concomitant increase in DNA damage and increases in inflammation-related cytokines. As there was an increase in DNA damage as well as expression of several EGF and TGF-ß pathway genes in response to inflammatory injury, we sought to determine if tumorigenesis was altered in the setting of inflammatory carcinogenesis. Se-deficient mice subjected to AOM/DSS treatment to model colitis-associated cancer (CAC) had increased tumor number, though not size, as well as increased incidence of high grade dysplasia. This increase in tumor initiation was likely due to a general increase in colonic DNA damage, as increased 8-OHdG staining was seen in Se-deficient tumors and adjacent, non-tumor mucosa. Taken together, our results indicate that Se deficiency worsens experimental colitis and promotes tumor development and progression in inflammatory carcinogenesis.


Asunto(s)
Carcinogénesis/metabolismo , Colitis/metabolismo , Neoplasias del Colon/metabolismo , Selenio/deficiencia , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Azoximetano , Carcinogénesis/inducido químicamente , Carcinogénesis/genética , Carcinogénesis/inmunología , Colitis/inducido químicamente , Colitis/genética , Colitis/inmunología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Daño del ADN , Desoxiguanosina/análogos & derivados , Desoxiguanosina/química , Sulfato de Dextran , Dieta , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/inmunología , Regulación de la Expresión Génica , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología , Pérdida de Peso
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA