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

Bases de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Carcinogenesis ; 39(9): 1176-1184, 2018 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-29901779

RESUMEN

Colorectal carcinogenesis is a progressive multistep process involving the sequential accumulation of genetic alterations in tumor suppressor genes and oncogenes. Downregulated by oncogenes 1 (Dro1/Ccdc80) has been shown to be a potent tumor suppressor of colorectal carcinogenesis in the genetic ApcMin/+ mouse model. In ApcMin/+ mice, loss of DRO1 strongly increases colonic tumor multiplicity and leads to the regular formation of adenocarcinoma in the colon. To investigate DRO1's role in chemically induced as well as inflammation-associated colorectal carcinogenesis, the effect of Dro1 inactivation was studied in mice subjected to the carcinogen azoxymethane (AOM) and upon combined treatment with AOM and the proinflammatory agent dextran sodium sulfate (DSS), respectively. Loss of DRO1 increases multiplicity of preneoplastic aberrant crypt foci and colonic tumors upon administration of AOM. Combined treatment with AOM and DSS leads to increased colonic tumor number and promotes formation of adenocarcinoma in the colon. Moreover, Dro1 inactivation aggravates histological signs of acute and chronic DSS-induced colitis, strongly enlarges the size of ulcerative lesions in the intestinal lining, and exacerbates clinical signs and morbidity by DSS. Our results demonstrate DRO1 to be a strong tumor suppressor in the chemically induced colon carcinogenic mouse model. Additionally, we demonstrate DRO1 to inhibit colitis-associated colon cancer formation and uncover a novel putative role for DRO1 in inflammatory bowel disease.


Asunto(s)
Adenocarcinoma/genética , Carcinogénesis/genética , Colitis/genética , Neoplasias Colorrectales/genética , Glicoproteínas/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Adenocarcinoma/patología , Animales , Azoximetano/toxicidad , Carcinógenos/toxicidad , Neoplasias Colorrectales/patología , Sulfato de Dextran/toxicidad , Transición Epitelial-Mesenquimal/genética , Proteínas de la Matriz Extracelular , Genes Supresores de Tumor , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
2.
Oncotarget ; 13: 615-627, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35422964

RESUMEN

Tumors are composed of the tumor cells and the surrounding microenvironment. Both are closely interwoven and interact by a complex and multifaceted cross-talk which plays an integral part in tumor initiation, growth, and progression. Dro1/Ccdc80 has been shown to be a potent suppressor of colorectal cancer and ubiquitous inactivation of Dro1/Ccdc80 strongly promoted colorectal carcinogenesis in ApcMin/+ mice and in a chemically-induced colorectal cancer model. The aim of the present study was to investigate whether Dro1/Ccdc80's tumor suppressive function is tumor-cell-autonomous. Expression of Dro1/Ccdc80 in cancer cells had no effect on both colon tumor development in ApcMin/+ mice and formation of xenograft tumors. In contrast, DRO1/CCDC80 loss in the microenvironment strongly increased tumor growth in xenograft models, inhibited cancer cell apoptosis, and promoted intestinal epithelial cell migration. Moreover, stromal Dro1/Ccdc80 inactivation facilitated formation of intestinal epithelial organoids. Expression analyses showed Dro1/Ccdc80 to be significantly down-regulated in murine gastric cancer associated fibroblasts, in ApcMin/+ colon tumor primary stromal cells and in microdissected stroma from human colorectal cancer compared to normal, non-tumor stroma. Our results demonstrate epithelial derived DRO1/CCDC80 to be dispensable for intestinal tissue homeostasis and identify Dro1/Ccdc80 as tumor suppressor in the tumor microenvironment.


Asunto(s)
Neoplasias del Colon , Proteínas de la Matriz Extracelular , Animales , Apoptosis , Carcinogénesis/genética , Neoplasias del Colon/patología , Proteínas de la Matriz Extracelular/genética , Genes Supresores de Tumor , Humanos , Ratones , Microambiente Tumoral
3.
Mol Cell Endocrinol ; 439: 286-296, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-27645901

RESUMEN

To investigate the role of DRO1 in obesity and adipogenesis in vivo, we generated a constitutive Dro1 knockout mouse model and analyzed the effect of DRO1 loss on body growth under standard and high fat diet feeding conditions. Loss of DRO1 resulted in a significant increase in body weight which was accompanied by a substantial expansion of white adipose tissue depots. The obese phenotype could be further enhanced by a high fat dietary challenge which also resulted in impaired glucose metabolism and the development of hepatosteatosis in Dro1 knockout mice. To study the role of DRO1 in adipocyte differentiation, primary stromal-vascular (SV) cells were isolated from inguinal white fat pads of knockout and control mice. In primary SV cells, depletion of DRO1 significantly promoted adipogenesis with upregulation of markers for adipogenesis (Cebpa, Pparg, Adipoq) and lipid metabolism (Dgat1, Dgat2). Our results demonstrate that DRO1 is a crucial regulator of energy homeostasis in vivo and functions as an inhibitor of adipogenesis in primary cells.


Asunto(s)
Adipocitos/patología , Diferenciación Celular , Glicoproteínas/deficiencia , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Obesidad/metabolismo , Obesidad/patología , Adipocitos/metabolismo , Adipogénesis , Tejido Adiposo , Animales , Biomarcadores/metabolismo , Peso Corporal , Dieta Alta en Grasa , Proteínas de la Matriz Extracelular , Hígado Graso/complicaciones , Hígado Graso/metabolismo , Hígado Graso/patología , Conducta Alimentaria , Femenino , Glucosa/metabolismo , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Ratones Noqueados , Obesidad/complicaciones , Tamaño de los Órganos , Células del Estroma/metabolismo , Regulación hacia Arriba
4.
Mol Cancer Res ; 12(11): 1655-62, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25053805

RESUMEN

UNLABELLED: Colorectal cancer develops from adenomatous precursor lesions by a multistep process that involves several independent mutational events in oncogenes and tumor suppressor genes. Inactivation of the adenomatous polyposis coli (APC) tumor suppressor gene is an early event and a prerequisite for the development of human colorectal adenoma. Previous in vitro studies identified DRO1 (CCDC80) to be a putative tumor suppressor gene that is negatively regulated in colorectal cancers and downregulated upon neoplastic transformation of epithelial cells. To investigate the in vivo role of DRO1 in colorectal carcinogenesis, a constitutive DRO1 knockout mouse model was generated. Disruption of DRO1 did not result in spontaneous intestinal tumor formation, consistent with the notion that DRO1 might have a role in suppressing the development of colon tumors in Apc(Min) (/+) mice, a widely used model for studying the role of APC in intestinal tumorigenesis that is hampered by the fact that mice predominantly develop adenomas in the small intestine and not in the colon. Here, deletion of DRO1 in Apc(Min) (/+) mice results in earlier death, a dramatically increased colonic tumor burden, and frequent development of colorectal carcinoma. Furthermore, enhanced phosphorylation of ERK1/2 is observed in colon epithelium and tumors from DRO1 knockout mice. Thus, this study reveals that inactivation of DRO1 is required for colorectal carcinogenesis in the Apc(Min) (/+) mouse and establishes a new mouse model for the study of colorectal cancer. IMPLICATIONS: This report characterizes a new mouse model for the study of colorectal cancer and establishes DRO1 (CCDC80) as a tumor suppressor via a mechanism involving ERK phosphorylation.


Asunto(s)
Poliposis Adenomatosa del Colon/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis , Proliferación Celular , Proteínas de la Matriz Extracelular , Femenino , Eliminación de Gen , Glicoproteínas/deficiencia , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Masculino , Ratones Noqueados , Proteínas Proto-Oncogénicas c-myc/metabolismo , Análisis de Supervivencia , Carga Tumoral , Proteínas Supresoras de Tumor/deficiencia , Vía de Señalización Wnt
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA