Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Oncogene ; 31(31): 3647-54, 2012 Aug 02.
Article in English | MEDLINE | ID: mdl-22120711

ABSTRACT

MafB, a member of the large Maf transcription factor family, is essential for the embryonic and terminal differentiation of pancreatic α- and ß-cells. However, the role of MafB in the control of adult islet-cell proliferation remains unknown. Considering its oncogenic potential in several other tissues, we investigated the possible alteration of its expression in adult mouse ß-cells under different conditions of proliferation. We found that MafB, in general silenced in these cells, was reexpressed in ∼30% of adaptive ß-cells both in gestational female mice and in mice fed with a high-fat diet. Importantly, reactivated MafB expression was also observed in the early ß-cell lesions and insulinomas that developed in ß-cell specific Men1 mutant mice, appearing in >80% of ß-cells in hyperplasic or dysplastic islets from the mutant mice >4 months of age. Moreover, MafB expression could be induced by glucose stimulation in INS-1 rat insulinoma cells. The induction was further reinforced following Men1 knockdown by siRNA. Furthermore, MafB overexpression in cultured ßTC3 cells enhanced cell foci formation both in culture medium and on soft agar, accompanied with the increased expression of Cyclin B1 and D2. Conversely, MafB downregulation by siRNA transfection reduced BrdU incorporation in INS-1E cells. Taken together, our data reveal that Men1 inactivation leads to MafB reexpression in mouse ß-cells in vivo, and provides evidence that deregulated ectopic MafB expression may have a hitherto unknown role in adult ß-cell proliferation and Men1-related tumorigenesis.


Subject(s)
Cell Proliferation , Insulin-Secreting Cells/metabolism , Insulinoma/metabolism , MafB Transcription Factor/biosynthesis , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins/biosynthesis , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cyclin B1/biosynthesis , Cyclin D2/biosynthesis , Diet, High-Fat , Female , Glucose/pharmacology , Insulinoma/pathology , Male , Mice , Mice, Inbred C57BL , Mutation , Pancreatic Neoplasms/pathology , Pregnancy , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/metabolism , Rats
2.
Endocr Relat Cancer ; 13(4): 1223-36, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17158767

ABSTRACT

Mutations of the MEN1 gene lead to the occurrence of multiple endocrine neoplasia type 1 (MEN1). To gain insights into the mechanisms of the tumorigenesis related to MEN1 inactivation, we have used mice in which the Men1 gene was specifically disrupted in pancreatic beta-cells. In these mice, we observed full penetrance of insulinoma with defined histological characteristics of tumorigenesis. To identify the genetic factors taking part in the tumour development, we performed gene expression profiling analysis of these insulinomas at different stages. Here, we show that in late stage insulinomas, 56 genes are up-regulated and 194 are down-regulated more than fourfold compared with normal pancreatic islets. Clustering analysis reveals the deregulation of Hox gene family and the genes involved in cell proliferation and cell cycle control. The altered expression of Igf2, Igfbp3 and Igfbp6 as well as cyclin A2, B2 and D2 are confirmed by quantitative RT-PCR, with the overexpression of all the three cyclins found in early stage insulinomas. Moreover, an increased proportion of cyclin A2- and D2-expressing cells and the overexpression of insulin-like growth factor 2 (IGF2) protein are detected in mouse Men1 insulinomas by immunostaining. Interestingly, the analysis of DNA methylation patterns by quantitative serial pyrosequencing reveals that four specific CpGs in the intragenic differentially methylated region 2 (DMR2) region of the Igf2 gene known to augment transcription through methylation are significantly hypermethylated in insulinomas of Men1 beta-cell mutant mice at 6 and 10 months of age, even before IGF2 overexpression can be detected. Thus, our data indicate the involvement of both genetic and epigenetic mechanisms in early tumorigenesis of beta-cells related to MEN1 inactivation.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Gene Expression Profiling , Insulin-Secreting Cells/metabolism , Insulinoma/genetics , Multiple Endocrine Neoplasia Type 1/genetics , Pancreatic Neoplasms/genetics , Animals , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Insulin-Secreting Cells/pathology , Insulinoma/metabolism , Insulinoma/pathology , Integrases/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Mice , Mice, Mutant Strains , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...