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.
Diabetologia ; 56(3): 542-52, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23238790

ABSTRACT

AIMS/HYPOTHESIS: The paucity of information on the epigenetic barriers that are blocking reprogramming protocols, and on what makes a beta cell unique, has hampered efforts to develop novel beta cell sources. Here, we aimed to identify enhancers in pancreatic islets, to understand their developmental ontologies, and to identify enhancers unique to islets to increase our understanding of islet-specific gene expression. METHODS: We combined H3K4me1-based nucleosome predictions with pancreatic and duodenal homeobox 1 (PDX1), neurogenic differentiation 1 (NEUROD1), v-Maf musculoaponeurotic fibrosarcoma oncogene family, protein A (MAFA) and forkhead box A2 (FOXA2) occupancy data to identify enhancers in mouse islets. RESULTS: We identified 22,223 putative enhancer loci in in vivo mouse islets. Our validation experiments suggest that nearly half of these loci are active in regulating islet gene expression, with the remaining regions probably poised for activity. We showed that these loci have at least nine developmental ontologies, and that islet enhancers predominately acquire H3K4me1 during differentiation. We next discriminated 1,799 enhancers unique to islets and showed that these islet-specific enhancers have reduced association with annotated genes, and identified a subset that are instead associated with novel islet-specific long non-coding RNAs (lncRNAs). CONCLUSIONS/INTERPRETATIONS: Our results indicate that genes with islet-specific expression and function tend to have enhancers devoid of histone methylation marks or, less often, that are bivalent or repressed, in embryonic stem cells and liver. Further, we identify a subset of enhancers unique to islets that are associated with novel islet-specific genes and lncRNAs. We anticipate that these data will facilitate the development of novel sources of functional beta cell mass.


Subject(s)
Islets of Langerhans/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromatin Immunoprecipitation , Enhancer Elements, Genetic/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Homeodomain Proteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Trans-Activators/metabolism
2.
Cell Death Dis ; 7: e2233, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27195679

ABSTRACT

Diabetes is a chronic disease that results from the body's inability to properly control circulating blood glucose levels. The loss of glucose homoeostasis can arise from a loss of ß-cell mass because of immune-cell-mediated attack, as in type 1 diabetes, and/or from dysfunction of individual ß-cells (in conjunction with target organ insulin resistance), as in type 2 diabetes. A better understanding of the transcriptional pathways regulating islet-cell survival is of great importance for the development of therapeutic strategies that target ß-cells for diabetes. To this end, we previously identified the transcription factor Myt3 as a pro-survival factor in islets following acute suppression of Myt3 in vitro. To determine the effects of Myt3 suppression on islet-cell survival in vivo, we used an adenovirus to express an shRNA targeting Myt3 in syngeneic optimal and marginal mass islet transplants, and demonstrate that suppression of Myt3 impairs the function of marginal mass grafts. Analysis of grafts 5 weeks post-transplant revealed that grafts transduced with the shMyt3 adenovirus contained ~20% the number of transduced cells as grafts transduced with a control adenovirus. In fact, increased apoptosis and significant cell loss in the shMyt3-transduced grafts was evident after only 5 days, suggesting that Myt3 suppression sensitizes islet cells to stresses present in the early post-transplant period. Specifically, we find that Myt3 suppression sensitizes islet cells to high glucose-induced cell death via upregulation of the pro-apoptotic Bcl2 family member Bim. Taken together these data suggest that Myt3 may be an important link between glucotoxic and immune signalling pathways.


Subject(s)
Bcl-2-Like Protein 11/genetics , Diabetes Mellitus, Experimental/genetics , Glucose/toxicity , Insulin-Secreting Cells/metabolism , Islets of Langerhans Transplantation , Transcription Factors/genetics , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Bcl-2-Like Protein 11/agonists , Bcl-2-Like Protein 11/metabolism , Cell Death/drug effects , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/mortality , Diabetes Mellitus, Experimental/therapy , Female , Gene Expression Regulation , Glucose/metabolism , Glucose Tolerance Test , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Mice , Mice, Inbred C57BL , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Streptozocin , Survival Analysis , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transplantation, Isogeneic
SELECTION OF CITATIONS
SEARCH DETAIL