Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 4 de 4
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Oncogene ; 38(25): 5107, 2019 Jun.
Article de Anglais | MEDLINE | ID: mdl-31068667

RÉSUMÉ

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.
Article de Anglais | MEDLINE | ID: mdl-30858547

RÉSUMÉ

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.


Sujet(s)
Adénocarcinome/génétique , Carcinogenèse/génétique , Colite/complications , Colite/génétique , Tumeurs du côlon/génétique , Protéines de répression/génétique , Facteurs de transcription/physiologie , Adénocarcinome/anatomopathologie , Animaux , Carcinogenèse/métabolisme , Carcinogenèse/anatomopathologie , Colite/anatomopathologie , Tumeurs du côlon/anatomopathologie , Femelle , Cellules HCT116 , Cellules HEK293 , Humains , Inflammation/complications , Inflammation/génétique , Inflammation/métabolisme , Inflammation/anatomopathologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Facteurs de transcription/génétique
3.
J Clin Invest ; 125(7): 2646-60, 2015 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-26053663

RÉSUMÉ

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.


Sujet(s)
Colite/complications , Tumeurs du côlon/étiologie , Sélénoprotéine P/physiologie , Animaux , Antioxydants/métabolisme , Apoptose , Tumeurs du côlon/anatomopathologie , Tumeurs du côlon/physiopathologie , Altération de l'ADN , Instabilité du génome , Haploinsuffisance , Macrophages/classification , Macrophages/anatomopathologie , Macrophages/physiologie , Souris , Souris de lignée C57BL , Souris knockout , Mutagenèse dirigée , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/physiologie , Stress oxydatif , Structure tertiaire des protéines , Sélénium/administration et posologie , Sélénium/métabolisme , Sélénoprotéine P/déficit , Sélénoprotéine P/génétique , Microenvironnement tumoral , Protéines suppresseurs de tumeurs/déficit , Protéines suppresseurs de tumeurs/génétique , Protéines suppresseurs de tumeurs/physiologie
4.
PLoS One ; 8(7): e67845, 2013.
Article de Anglais | MEDLINE | ID: mdl-23861820

RÉSUMÉ

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.


Sujet(s)
Carcinogenèse/métabolisme , Colite/métabolisme , Tumeurs du côlon/métabolisme , Sélénium/déficit , 8-Hydroxy-2'-désoxyguanosine , Animaux , Oxyde de diméthyl-diazène , Carcinogenèse/induit chimiquement , Carcinogenèse/génétique , Carcinogenèse/immunologie , Colite/induit chimiquement , Colite/génétique , Colite/immunologie , Tumeurs du côlon/induit chimiquement , Tumeurs du côlon/génétique , Tumeurs du côlon/immunologie , Altération de l'ADN , Désoxyguanosine/analogues et dérivés , Désoxyguanosine/composition chimique , Sulfate dextran , Régime alimentaire , Facteur de croissance épidermique/génétique , Facteur de croissance épidermique/immunologie , Régulation de l'expression des gènes , Inflammation/induit chimiquement , Inflammation/génétique , Inflammation/immunologie , Inflammation/métabolisme , Souris , Souris de lignée C57BL , Transduction du signal , Facteur de croissance transformant bêta/génétique , Facteur de croissance transformant bêta/immunologie , Perte de poids
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE