Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 18 de 18
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Gastroenterology ; 160(5): 1694-1708.e3, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33388316

RÉSUMÉ

BACKGROUND & AIMS: Patients with inflammatory bowel disease (IBD) demonstrate nutritional selenium deficiencies and are at greater risk of developing colon cancer. Previously, we determined that global reduction of the secreted antioxidant selenium-containing protein, selenoprotein P (SELENOP), substantially increased tumor development in an experimental colitis-associated cancer (CAC) model. We next sought to delineate tissue-specific contributions of SELENOP to intestinal inflammatory carcinogenesis and define clinical context. METHODS: Selenop floxed mice crossed with Cre driver lines to delete Selenop from the liver, myeloid lineages, or intestinal epithelium were placed on an azoxymethane/dextran sodium sulfate experimental CAC protocol. SELENOP loss was assessed in human ulcerative colitis (UC) organoids, and expression was queried in human and adult UC samples. RESULTS: Although large sources of SELENOP, both liver- and myeloid-specific Selenop deletion failed to modify azoxymethane/dextran sodium sulfate-mediated tumorigenesis. Instead, epithelial-specific deletion increased CAC tumorigenesis, likely due to elevated oxidative stress with a resulting increase in genomic instability and augmented tumor initiation. SELENOP was down-regulated in UC colon biopsies and levels were inversely correlated with endoscopic disease severity and tissue S100A8 (calprotectin) gene expression. CONCLUSIONS: Although global selenium status is typically assessed by measuring liver-derived plasma SELENOP levels, our results indicate that the peripheral SELENOP pool is dispensable for CAC. Colonic epithelial SELENOP is the main contributor to local antioxidant capabilities. Thus, colonic SELENOP is the most informative means to assess selenium levels and activity in IBD patients and may serve as a novel biomarker for UC disease severity and identify patients most predisposed to CAC development.


Sujet(s)
Rectocolite hémorragique/métabolisme , Néoplasmes associés aux colites/prévention et contrôle , Colite/métabolisme , Côlon/métabolisme , Muqueuse intestinale/métabolisme , Stress oxydatif , Sélénoprotéine P/métabolisme , Adolescent , Animaux , Oxyde de diméthyl-diazène , Études cas-témoins , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Enfant , Enfant d'âge préscolaire , Colite/induit chimiquement , Colite/génétique , Rectocolite hémorragique/génétique , Néoplasmes associés aux colites/induit chimiquement , Néoplasmes associés aux colites/génétique , Néoplasmes associés aux colites/métabolisme , Côlon/anatomopathologie , Altération de l'ADN , Sulfate dextran , Modèles animaux de maladie humaine , Femelle , Instabilité du génome , Humains , Muqueuse intestinale/anatomopathologie , Foie/métabolisme , Mâle , Souris knockout , Cellules myéloïdes/métabolisme , Sélénoprotéine P/génétique
2.
NPJ Parkinsons Dis ; 6(1): 34, 2020 Nov 13.
Article de Anglais | MEDLINE | ID: mdl-33298952

RÉSUMÉ

The catecholamine neurotransmitter dopamine has the potential to act as an endogenous neurotoxin when its vesicular sequestration is dysregulated. Despite postmortem analyses from patients with Parkinson's disease that demonstrate decreased vesicular sequestration of dopamine with a corresponding increase in dopamine metabolism, dopamine's contribution to nigrostriatal dopaminergic degeneration in Parkinson's disease has been debated. Here, we present a new in vivo model demonstrating the induction of Parkinson's disease-associated pathogenic mechanisms of degeneration resulting from acquired dysregulation of dopamine sequestration in nigrostriatal dopaminergic neurons in adult rats. Utilizing adeno-associated virus (serotype 2), viral-mediated small-hairpin RNA interference of endogenous vesicular monoamine transporter 2 (VMAT2) expression resulted in a loss of VMAT2 protein expression in transduced dopaminergic cell bodies in the substantia nigra with a corresponding loss of VMAT2 protein within the striatal terminals. The loss of VMAT2 resulted in an accumulation of cytosolic dopamine and subsequent increased dopamine metabolism, deficits in dopamine-mediated behaviors, and degeneration of nigrostriatal dopaminergic neurons that was rescued through reintroduction of exogenous VMAT2, demonstrating that the toxicity was specific to the loss of VMAT2. Analysis of parkinsonian pathogenic mechanisms of degeneration identified oxidative damage, activation of Parkinson's disease-associated kinase LRRK2, and the formation of aberrant α-synuclein. This model demonstrates that a progressive acquired loss of VMAT2 expression in adulthood is sufficient to induce Parkinson's disease-associated pathogenic mechanisms of degeneration and provides a new model to further investigate the consequences of cytosolic dopamine.

3.
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.

4.
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
5.
Mucosal Immunol ; 11(5): 1363-1374, 2018 09.
Article de Anglais | MEDLINE | ID: mdl-29907869

RÉSUMÉ

Blood vessel epicardial substance (BVES), or POPDC1, is a tight junction-associated transmembrane protein that modulates epithelial-to-mesenchymal transition (EMT) via junctional signaling pathways. There have been no in vivo studies investigating the role of BVES in colitis. We hypothesized that BVES is critical for maintaining colonic epithelial integrity. At baseline, Bves-/- mouse colons demonstrate increased crypt height, elevated proliferation, decreased apoptosis, altered intestinal lineage allocation, and dysregulation of tight junctions with functional deficits in permeability and altered intestinal immunity. Bves-/- mice inoculated with Citrobacter rodentium had greater colonic injury, increased colonic and mesenteric lymph node bacterial colonization, and altered immune responses after infection. We propose that increased bacterial colonization and translocation result in amplified immune responses and worsened injury. Similarly, dextran sodium sulfate (DSS) treatment resulted in greater histologic injury in Bves-/- mice. Two different human cell lines (Caco2 and HEK293Ts) co-cultured with enteropathogenic E. coli showed increased attaching/effacing lesions in the absence of BVES. Finally, BVES mRNA levels were reduced in human ulcerative colitis (UC) biopsy specimens. Collectively, these studies suggest that BVES plays a protective role both in ulcerative and infectious colitis and identify BVES as a critical protector of colonic mucosal integrity.


Sujet(s)
Rectocolite hémorragique/métabolisme , Côlon/métabolisme , Cellules épithéliales/métabolisme , Absorption intestinale/physiologie , Protéines membranaires/métabolisme , Adulte , Animaux , Cellules Caco-2 , Molécules d'adhérence cellulaire , Lignée cellulaire , Lignée cellulaire tumorale , Citrobacter rodentium/pathogénicité , Techniques de coculture , Côlon/effets des médicaments et des substances chimiques , Sulfate dextran/pharmacologie , Cellules épithéliales/effets des médicaments et des substances chimiques , Escherichia coli/métabolisme , Femelle , Cellules HEK293 , Humains , Absorption intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/métabolisme , Mâle , Souris , Souris de lignée C57BL , Adulte d'âge moyen , Protéines du muscle , Perméabilité/effets des médicaments et des substances chimiques , ARN messager/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/physiologie , Jonctions serrées/effets des médicaments et des substances chimiques , Jonctions serrées/métabolisme
6.
JCI Insight ; 2(16)2017 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-28814670

RÉSUMÉ

MTG16 is a member of the myeloid translocation gene (MTG) family of transcriptional corepressors. While MTGs were originally identified in chromosomal translocations in acute myeloid leukemia, recent studies have uncovered a role in intestinal biology. For example, Mtg16-/- mice have increased intestinal proliferation and are more sensitive to intestinal injury in colitis models. MTG16 is also underexpressed in patients with moderate/severe ulcerative colitis. Based on these findings, we postulated that MTG16 might protect against colitis-associated carcinogenesis. MTG16 was downregulated at the protein and RNA levels in patients with inflammatory bowel disease and in those with colitis-associated carcinoma. Mtg16-/- mice subjected to inflammatory carcinogenesis modeling exhibited worse colitis and increased tumor multiplicity and size. Loss of MTG16 also increased severity of dysplasia, apoptosis, proliferation, DNA damage, and WNT signaling. Moreover, transplantation of WT marrow into Mtg16-/- mice failed to rescue the Mtg16-/- protumorigenic phenotypes, indicating an epithelium-specific role for MTG16. While MTG dysfunction is widely appreciated in hematopoietic malignancies, the role of this gene family in epithelial homeostasis, and in colon cancer, was unrealized. This report identifies MTG16 as an important modulator of colitis and tumor development in inflammatory carcinogenesis.

7.
Gut ; 66(5): 852-862, 2017 05.
Article de Anglais | MEDLINE | ID: mdl-28389570

RÉSUMÉ

OBJECTIVE: Blood vessel epicardial substance (BVES) is a tight junction-associated protein that regulates epithelial-mesenchymal states and is underexpressed in epithelial malignancy. However, the functional impact of BVES loss on tumourigenesis is unknown. Here we define the in vivo role of BVES in colitis-associated cancer (CAC), its cellular function and its relevance to patients with IBD. DESIGN: We determined BVES promoter methylation status using an Infinium HumanMethylation450 array screen of patients with UC with and without CAC. We also measured BVES mRNA levels in a tissue microarray consisting of normal colons and CAC samples. Bves-/- and wild-type mice (controls) were administered azoxymethane (AOM) and dextran sodium sulfate (DSS) to induce tumour formation. Last, we used a yeast two-hybrid screen to identify BVES interactors and performed mechanistic studies in multiple cell lines to define how BVES reduces c-Myc levels. RESULTS: BVES mRNA was reduced in tumours from patients with CAC via promoter hypermethylation. Importantly, BVES promoter hypermethylation was concurrently present in distant non-malignant-appearing mucosa. As seen in human patients, Bves was underexpressed in experimental inflammatory carcinogenesis, and Bves-/- mice had increased tumour multiplicity and degree of dysplasia after AOM/DSS administration. Molecular analysis of Bves-/- tumours revealed Wnt activation and increased c-Myc levels. Mechanistically, we identified a new signalling pathway whereby BVES interacts with PR61α, a protein phosphatase 2A regulatory subunit, to mediate c-Myc destruction. CONCLUSION: Loss of BVES promotes inflammatory tumourigenesis through dysregulation of Wnt signalling and the oncogene c-Myc. BVES promoter methylation status may serve as a CAC biomarker.


Sujet(s)
Carcinogenèse/génétique , Molécules d'adhérence cellulaire/génétique , Rectocolite hémorragique/métabolisme , Tumeurs du côlon/métabolisme , Protéines membranaires/génétique , Protéines du muscle/génétique , Protéines proto-oncogènes c-myc/métabolisme , Animaux , Marqueurs biologiques tumoraux/génétique , Cellules Caco-2 , Colite/induit chimiquement , Colite/génétique , Colite/métabolisme , Rectocolite hémorragique/génétique , Côlon/métabolisme , Tumeurs du côlon/génétique , Tumeurs du côlon/anatomopathologie , Méthylation de l'ADN , Sulfate dextran , Régulation négative , Femelle , Analyse de profil d'expression de gènes , Cellules HEK293 , Humains , Mâle , Souris , Souris knockout , Régions promotrices (génétique) , Protein Phosphatase 2/métabolisme , Protéines proto-oncogènes c-myc/génétique , ARN messager/métabolisme , Voie de signalisation Wnt
8.
Cell Mol Life Sci ; 74(4): 607-616, 2017 02.
Article de Anglais | MEDLINE | ID: mdl-27563706

RÉSUMÉ

Selenium is an essential micronutrient that is incorporated into at least 25 selenoproteins encoded by the human genome, many of which serve antioxidant functions. Because patients with inflammatory bowel disease (IBD) demonstrate nutritional deficiencies and are at increased risk for colon cancer due to heightened inflammation and oxidative stress, selenoprotein dysfunction may contribute to disease progression. Over the years, numerous studies have analyzed the effects of selenoprotein loss and shown that they are important mediators of intestinal inflammation and carcinogenesis. In particular, recent work has focused on the role of selenoprotein P (SEPP1), a major selenium transport protein which also has endogenous antioxidant function. These experiments determined SEPP1 loss altered immune and epithelial cellular function in a murine model of colitis-associated carcinoma. Here, we discuss the current knowledge of SEPP1 and selenoprotein function in the setting of IBD, colitis, and inflammatory tumorigenesis.


Sujet(s)
Carcinogenèse/immunologie , Colite/immunologie , Tumeurs du côlon/immunologie , Maladies inflammatoires intestinales/immunologie , Stress oxydatif , Sélénium/immunologie , Sélénoprotéines/immunologie , Animaux , Carcinogenèse/métabolisme , Carcinogenèse/anatomopathologie , Colite/complications , Colite/métabolisme , Colite/anatomopathologie , Côlon/immunologie , Côlon/métabolisme , Côlon/anatomopathologie , Tumeurs du côlon/étiologie , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Glutathione peroxidase/immunologie , Glutathione peroxidase/métabolisme , Humains , Maladies inflammatoires intestinales/complications , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/anatomopathologie , Sélénium/métabolisme , Sélénoprotéine P/immunologie , Sélénoprotéine P/métabolisme , Sélénoprotéines/métabolisme
9.
Methods Mol Biol ; 1422: 297-307, 2016.
Article de Anglais | MEDLINE | ID: mdl-27246042

RÉSUMÉ

Our understanding of colitis-associated carcinoma (CAC) has benefited substantially from mouse models that faithfully recapitulate human CAC. Chemical models, in particular, have enabled fast and efficient analysis of genetic and environmental modulators of CAC without the added requirement of time-intensive genetic crossings. Here we describe the Azoxymethane (AOM)/Dextran Sodium Sulfate (DSS) mouse model of inflammatory colorectal cancer.


Sujet(s)
Oxyde de diméthyl-diazène , Colite/induit chimiquement , Colite/anatomopathologie , Tumeurs colorectales/induit chimiquement , Tumeurs colorectales/anatomopathologie , Sulfate dextran , Modèles animaux de maladie humaine , Animaux , Colite/complications , Humains , Souris
10.
Sci Transl Med ; 8(342): 342ra78, 2016 06 08.
Article de Anglais | MEDLINE | ID: mdl-27280685

RÉSUMÉ

α-Synuclein accumulation and mitochondrial dysfunction have both been strongly implicated in the pathogenesis of Parkinson's disease (PD), and the two appear to be related. Mitochondrial dysfunction leads to accumulation and oligomerization of α-synuclein, and increased levels of α-synuclein cause mitochondrial impairment, but the basis for this bidirectional interaction remains obscure. We now report that certain posttranslationally modified species of α-synuclein bind with high affinity to the TOM20 (translocase of the outer membrane 20) presequence receptor of the mitochondrial protein import machinery. This binding prevented the interaction of TOM20 with its co-receptor, TOM22, and impaired mitochondrial protein import. Consequently, there were deficient mitochondrial respiration, enhanced production of reactive oxygen species, and loss of mitochondrial membrane potential. Examination of postmortem brain tissue from PD patients revealed an aberrant α-synuclein-TOM20 interaction in nigrostriatal dopaminergic neurons that was associated with loss of imported mitochondrial proteins, thereby confirming this pathogenic process in the human disease. Modest knockdown of endogenous α-synuclein was sufficient to maintain mitochondrial protein import in an in vivo model of PD. Furthermore, in in vitro systems, overexpression of TOM20 or a mitochondrial targeting signal peptide had beneficial effects and preserved mitochondrial protein import. This study characterizes a pathogenic mechanism in PD, identifies toxic species of wild-type α-synuclein, and reveals potential new therapeutic strategies for neuroprotection.


Sujet(s)
Protéines mitochondriales/métabolisme , Maladie de Parkinson/métabolisme , Récepteurs cytoplasmiques et nucléaires/métabolisme , alpha-Synucléine/métabolisme , Animaux , Protéines de transport membranaire/génétique , Protéines de transport membranaire/métabolisme , Mitochondries/métabolisme , Protéines du complexe d'import des protéines précurseurs mitochondriales , Protéines mitochondriales/génétique , Maladie de Parkinson/génétique , Liaison aux protéines , Transport des protéines/génétique , Transport des protéines/physiologie , Rats , Souches mutantes de rat , Récepteurs de surface cellulaire , Récepteurs cytoplasmiques et nucléaires/génétique , alpha-Synucléine/génétique
11.
Stem Cells ; 34(6): 1626-36, 2016 06.
Article de Anglais | MEDLINE | ID: mdl-26891025

RÉSUMÉ

Blood vessel epicardial substance (BVES/Popdc1) is a junctional-associated transmembrane protein that is underexpressed in a number of malignancies and regulates epithelial-to-mesenchymal transition. We previously identified a role for BVES in regulation of the Wnt pathway, a modulator of intestinal stem cell programs, but its role in small intestinal (SI) biology remains unexplored. We hypothesized that BVES influences intestinal stem cell programs and is critical to SI homeostasis after radiation injury. At baseline, Bves(-/-) mice demonstrated increased crypt height, as well as elevated proliferation and expression of the stem cell marker Lgr5 compared to wild-type (WT) mice. Intercross with Lgr5-EGFP reporter mice confirmed expansion of the stem cell compartment in Bves(-/-) mice. To examine stem cell function after BVES deletion, we used ex vivo 3D-enteroid cultures. Bves(-/-) enteroids demonstrated increased stemness compared to WT, when examining parameters such as plating efficiency, stem spheroid formation, and retention of peripheral cystic structures. Furthermore, we observed increased proliferation, expression of crypt-base columnar "CBC" and "+4" stem cell markers, amplified Wnt signaling, and responsiveness to Wnt activation in the Bves(-/-) enteroids. Bves expression was downregulated after radiation in WT mice. Moreover, after radiation, Bves(-/-) mice demonstrated significantly greater SI crypt viability, proliferation, and amplified Wnt signaling in comparison to WT mice. Bves(-/-) mice also demonstrated elevations in Lgr5 and Ascl2 expression, and putative damage-responsive stem cell populations marked by Bmi1 and TERT. Therefore, BVES is a key regulator of intestinal stem cell programs and mucosal homeostasis. Stem Cells 2016;34:1626-1636.


Sujet(s)
Molécules d'adhérence cellulaire/métabolisme , Rayons gamma , Intestins/cytologie , Protéines du muscle/métabolisme , Cellules souches/cytologie , Animaux , Molécules d'adhérence cellulaire/génétique , Survie cellulaire/effets des radiations , Régulation négative/effets des radiations , Femelle , Délétion de gène , Homéostasie/effets des radiations , Mâle , Souris de lignée C57BL , Protéines du muscle/génétique , Radiotolérance/effets des radiations , Sphéroïdes de cellules/métabolisme , Sphéroïdes de cellules/effets des radiations , Cellules souches/métabolisme , Cellules souches/effets des radiations , Voie de signalisation Wnt/effets des radiations
12.
Bio Protoc ; 6(12)2016 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-29333475

RÉSUMÉ

Small intestinal organoids, otherwise known as enteroids, have become an increasingly utilized model for intestinal biology in vitro as they recapitulate the various epithelial cells within the intestinal crypt (Mahe et al., 2013; Sato et al., 2009). Assessment of growth dynamics within these cultures is an important step to understanding how alterations in gene expression, treatment with protective and toxic agents, and genetic mutations alter properties essential for crypt growth and survival as well as the stem cell properties of the individual cells within the crypt. This protocol describes a method of visualization of proliferating cells within the crypt in three dimensions (Barrett et al., 2015). Whole-mount proliferation staining of enteroids using EdU incorporation enables the researcher to view all proliferating cells within the enteroid as opposed to obtaining growth information in thin slices as would be seen with embedding and sectioning, ensuring a true representation of proliferation from the stem cell compartment to the terminally differentiated cells of the crypt.

13.
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
14.
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
15.
Cancer Res ; 73(3): 1245-55, 2013 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-23221387

RÉSUMÉ

The glutathione peroxidases, a family of selenocysteine-containing redox enzymes, play pivotal roles in balancing the signaling, immunomodulatory, and deleterious effects of reactive oxygen species (ROS). The glutathione peroxidase GPX3 is the only extracellular member of this family, suggesting it may defend cells against ROS in the extracellular environment. Notably, GPX3 hypermethylation and underexpression occur commonly in prostate, gastric, cervical, thyroid, and colon cancers. We took a reverse genetics approach to investigate whether GPX3 would augment inflammatory colonic tumorigenesis, a process characterized by oxidative stress and inflammation, comparing Gpx3(-/-) mice in an established two-stage model of inflammatory colon carcinogenesis. Gpx3-deficient mice exhibited an increased tumor number, though not size, along with a higher degree of dysplasia. In addition, they exhibited increased inflammation with redistribution toward protumorigenic M2 macrophage subsets, increased proliferation, hyperactive WNT signaling, and increased DNA damage. To determine the impact of acute gene loss in an established colon cancer line, we silenced GPX3 in human Caco2 cells, resulting in increased ROS production, DNA damage and apoptosis in response to oxidative stress, combined with decreased contact-independent growth. Taken together, our results suggested an immunomodulatory role for GPX3 that limits the development of colitis-associated carcinoma.


Sujet(s)
Colite/complications , Tumeurs du côlon/étiologie , Glutathione peroxidase/physiologie , Protéines suppresseurs de tumeurs/physiologie , Animaux , Cellules Caco-2 , Altération de l'ADN , Humains , Souris , Souris de lignée C57BL , Stress oxydatif
16.
PLoS One ; 7(12): e51205, 2012.
Article de Anglais | MEDLINE | ID: mdl-23251453

RÉSUMÉ

Myeloid translocation genes (MTGs) are transcriptional corepressors originally identified in acute myelogenous leukemia that have recently been linked to epithelial malignancy with non-synonymous mutations identified in both MTG8 and MTG16 in colon, breast, and lung carcinoma in addition to functioning as negative regulators of WNT and Notch signaling. A yeast two-hybrid approach was used to discover novel MTG binding partners. This screen identified the Zinc fingers, C2H2 and BTB domain containing (ZBTB) family members ZBTB4 and ZBTB38 as MTG16 interacting proteins. ZBTB4 is downregulated in breast cancer and modulates p53 responses. Because ZBTB33 (Kaiso), like MTG16, modulates Wnt signaling at the level of TCF4, and its deletion suppresses intestinal tumorigenesis in the Apc(Min) mouse, we determined that Kaiso also interacted with MTG16 to modulate transcription. The zinc finger domains of Kaiso as well as ZBTB4 and ZBTB38 bound MTG16 and the association with Kaiso was confirmed using co-immunoprecipitation. MTG family members were required to efficiently repress both a heterologous reporter construct containing Kaiso binding sites (4×KBS) and the known Kaiso target, Matrix metalloproteinase-7 (MMP-7/Matrilysin). Moreover, chromatin immunoprecipitation studies placed MTG16 in a complex occupying the Kaiso binding site on the MMP-7 promoter. The presence of MTG16 in this complex, and its contributions to transcriptional repression both required Kaiso binding to its binding site on DNA, establishing MTG16-Kaiso binding as functionally relevant in Kaiso-dependent transcriptional repression. Examination of a large multi-stage CRC expression array dataset revealed patterns of Kaiso, MTG16, and MMP-7 expression supporting the hypothesis that loss of either Kaiso or MTG16 can de-regulate a target promoter such as that of MMP-7. These findings provide new insights into the mechanisms of transcriptional control by ZBTB family members and broaden the scope of co-repressor functions for the MTG family, suggesting coordinate regulation of transcription by Kaiso/MTG complexes in cancer.


Sujet(s)
Régions promotrices (génétique) , Protéines de répression/métabolisme , Facteurs de transcription/physiologie , Transcription génétique , Protéines suppresseurs de tumeurs/métabolisme , Sites de fixation , Immunoprécipitation de la chromatine , Technique d'immunofluorescence , Techniques de knock-down de gènes , Cellules HEK293 , Cellules HT29 , Humains , Cellules K562 , Matrix metalloproteinase 7/génétique , Séquençage par oligonucléotides en batterie , Facteurs de transcription/génétique
17.
J Clin Invest ; 121(10): 4056-69, 2011 Oct.
Article de Anglais | MEDLINE | ID: mdl-21911938

RÉSUMÉ

The acquisition of a mesenchymal phenotype is a critical step in the metastatic progression of epithelial carcinomas. Adherens junctions (AJs) are required for suppressing this epithelial-mesenchymal transition (EMT) but less is known about the role of tight junctions (TJs) in this process. Here, we investigated the functions of blood vessel epicardial substance (BVES, also known as POPDC1 and POP1), an integral membrane protein that regulates TJ formation. BVES was found to be underexpressed in all stages of human colorectal carcinoma (CRC) and in adenomatous polyps, indicating its suppression occurs early in transformation. Similarly, the majority of CRC cell lines tested exhibited decreased BVES expression and promoter DNA hypermethylation, a modification associated with transcriptional silencing. Treatment with a DNA-demethylating agent restored BVES expression in CRC cell lines, indicating that methylation represses BVES expression. Reexpression of BVES in CRC cell lines promoted an epithelial phenotype, featuring decreased proliferation, migration, invasion, and anchorage-independent growth; impaired growth of an orthotopic xenograft; and blocked metastasis. Conversely, interfering with BVES function by expressing a dominant-negative mutant in human corneal epithelial cells induced mesenchymal features. These biological outcomes were associated with changes in AJ and TJ composition and related signaling. Therefore, BVES prevents EMT, and its epigenetic silencing may be an important step in promoting EMT programs during colon carcinogenesis.


Sujet(s)
Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/physiopathologie , Transition épithélio-mésenchymateuse/physiologie , Épithélium antérieur de la cornée/cytologie , Épithélium antérieur de la cornée/physiologie , Protéines membranaires/physiologie , Adénocarcinome/génétique , Adénocarcinome/anatomopathologie , Adénocarcinome/physiopathologie , Jonctions adhérentes/anatomopathologie , Jonctions adhérentes/physiologie , Animaux , Molécules d'adhérence cellulaire , Lignée cellulaire tumorale , Tumeurs du côlon/génétique , Tumeurs du côlon/anatomopathologie , Tumeurs du côlon/physiopathologie , Polypes coliques/génétique , Polypes coliques/anatomopathologie , Polypes coliques/physiopathologie , Tumeurs colorectales/génétique , Méthylation de l'ADN , Transition épithélio-mésenchymateuse/génétique , Expression des gènes , Extinction de l'expression des gènes , Humains , Protéines membranaires/génétique , Souris , Souris nude , Protéines du muscle , Mutation , Transplantation tumorale , Régions promotrices (génétique) , Transduction du signal , Jonctions serrées/anatomopathologie , Jonctions serrées/physiologie , Transplantation hétérologue
18.
Cancer Res ; 71(4): 1302-12, 2011 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-21303973

RÉSUMÉ

Myeloid Translocation Gene, Related-1 (MTGR1) CBFA2T2 is a member of the Myeloid Translocation Gene (MTG) family of transcriptional corepressors. The remaining two family members, MTG8 (RUNX1T1) and MTG16 (CBFA2T3) are identified as targets of chromosomal translocations in acute myeloid leukemia (AML). Mtgr1(-/-) mice have defects in intestinal lineage allocation and wound healing. Moreover, these mice show signs of impaired intestinal stem cell function. Based on these phenotypes, we hypothesized that MTGR1 may influence tumorigenesis arising in an inflammatory background. We report that Mtgr1(-/-) mice were protected from tumorigenesis when injected with azoxymethane (AOM) and then subjected to repeated cycles of dextran sodium sulfate (DSS). Tumor cell proliferation was comparable, but Mtgr1(-/-) tumors had significantly higher apoptosis rates. These phenotypes were dependent on epithelial injury, the resultant inflammation, or a combination of both as there was no difference in aberrant crypt foci (ACF) or tumor burden when animals were treated with AOM as the sole agent. Gene expression analysis indicated that Mtgr1(-/-) tumors had significant upregulation of inflammatory networks, and immunohistochemistry (IHC) for immune cell subsets revealed a marked multilineage increase in infiltrates, consisting predominately of CD3(+) and natural killer T (NKT) cells as well as macrophages. Transplantation of wild type (WT) bone marrow into Mtgr1(-/-) mice, and the reciprocal transplant, did not alter the phenotype, ruling out an MTGR1 hematopoietic cell-autonomous mechanism. Our findings indicate that MTGR1 is required for efficient inflammatory carcinogenesis in this model, and implicate its dysfunction in colitis-associated carcinoma. This represents the first report functionally linking MTGR1 to intestinal tumorigenesis.


Sujet(s)
Carcinomes/génétique , Transformation cellulaire néoplasique/génétique , Colite/anatomopathologie , Tumeurs du côlon/génétique , Protéines de répression/physiologie , Animaux , Oxyde de diméthyl-diazène , Carcinomes/anatomopathologie , Transformation cellulaire néoplasique/effets des médicaments et des substances chimiques , Colite/induit chimiquement , Colite/génétique , Tumeurs du côlon/anatomopathologie , Sulfate dextran , Modèles animaux de maladie humaine , Évolution de la maladie , Régulation de l'expression des gènes tumoraux , Cellules HCT116 , Humains , Souris , Souris de lignée C57BL , Souris knockout , Protéines de répression/génétique , Protéines de répression/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE