Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 10(10): e0141470, 2015.
Article in English | MEDLINE | ID: mdl-26505193

ABSTRACT

To better understand how ß-cells respond to proinflammatory cytokines we mapped the locations of histone 3 lysine 4 monomethylation (H3K4me1), a post-translational histone modification enriched at active and poised cis-regulatory regions, in IFNγ, Il-1ß, and TNFα treated pancreatic islets. We identified 96,721 putative cis-regulatory loci, of which 3,590 were generated de novo, 3,204 had increased H3K4me1, and 5,354 had decreased H3K4me1 in IFNγ, Il-1ß, and TNFα exposed islets. Roughly 10% of the de novo and increased regions were enriched for the repressive histone modification histone 3 lysine 27 trimethylation (H3K27me3) in untreated cells, and these were frequently associated with chemokine genes. We show that IFNγ, Il-1ß, and TNFα exposure overcomes this repression and induces chemokine gene activation in as little as three hours, and that this expression persists for days in absence of continued IFNγ, Il-1ß, and TNFα exposure. We implicate trithorax group (TrxG) complexes as likely players in the conversion of these repressed loci to an active state. To block the activity of these complexes, we suppressed Wdr5, a core component of the TrxG complexes, and used the H3K27me3 demethylase inhibitor GSK-J4. We show that GSK-J4 is particularly effective in blunting IFNγ, Il-1ß, and TNFα-induced chemokine gene expression in ß-cells; however, it induced significant islet-cell apoptosis and ß-cell dysfunction. Wdr5 suppression also reduced IFNγ, Il-1ß, and TNFα induced chemokine gene expression in ß-cells without affecting islet-cell survival or ß-cell function after 48hrs, but did begin to increase islet-cell apoptosis and ß-cell dysfunction after four days of treatment. Taken together these data suggest that the TrxG complex is potentially a viable target for preventing cytokine induced chemokine gene expression in ß-cells.


Subject(s)
Histones/genetics , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Proteins/metabolism , Adenoviridae/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Benzazepines/pharmacology , Gene Expression Regulation/drug effects , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/biosynthesis , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Insulin-Secreting Cells/drug effects , Interferon-gamma/administration & dosage , Interleukin-1beta/administration & dosage , Intracellular Signaling Peptides and Proteins , Islets of Langerhans/drug effects , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Myeloid-Lymphoid Leukemia Protein/chemistry , Myeloid-Lymphoid Leukemia Protein/genetics , Proteins/genetics , Pyrimidines/pharmacology , Tumor Necrosis Factor-alpha/administration & dosage
2.
Mol Endocrinol ; 29(9): 1254-68, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26177052

ABSTRACT

Myt3 is a prosurvival factor in pancreatic islets; however, its role in islet-cell development is not known. Here, we demonstrate that myelin transcription factor 3 (Myt3) is expressed in migrating islet cells in the developing and neonatal pancreas and thus sought to determine whether Myt3 plays a role in this process. Using an ex vivo model of islet-cell migration, we demonstrate that Myt3 suppression significantly inhibits laminin-V/integrin-ß1-dependent α- and ß-cell migration onto 804G, and impaired 804G-induced F-actin and E-cadherin redistribution. Exposure of islets to proinflammatory cytokines, which suppress Myt3 expression, had a similar effect, whereas Myt3 overexpression partially rescued the migratory ability of the islet cells. We show that loss of islet-cell migration, due to Myt3 suppression or cytokine exposure, is independent of effects on islet-cell survival or proliferation. Myt3 suppression also had no effect on glucose-induced calcium influx, F-actin remodeling or insulin secretion by ß-cells. RNA-sequencing (RNA-seq) analysis of transduced islets showed that Myt3 suppression results in the up-regulation of Tgfbi, a secreted diabetogenic factor thought to impair cellular adhesion. Exposure of islets to exogenous transforming growth factor ß-induced (Tgfbi) impaired islet-cell migration similar to Myt3 suppression. Taken together, these data suggest a model by which cytokine-induced Myt3 suppression leads to Tgfbi de-repression and subsequently to impaired islet-cell migration, revealing a novel role for Myt3 in regulating islet-cell migration.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Movement/physiology , Insulin-Secreting Cells/metabolism , Integrin beta1/metabolism , Transcription Factors/metabolism , Actins/metabolism , Animals , Cadherins/metabolism , Calcium/metabolism , Cell Adhesion/physiology , Cell Proliferation/physiology , Cytokines/pharmacology , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/pharmacology , Female , Insulin/metabolism , Insulin Secretion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Transcription Factors/antagonists & inhibitors , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/pharmacology , Kalinin
3.
PLoS One ; 7(12): e51501, 2012.
Article in English | MEDLINE | ID: mdl-23236509

ABSTRACT

AIMS/HYPOTHESIS: We previously identified the transcription factor Myt3 as specifically expressed in pancreatic islets. Here, we sought to determine the expression and regulation of Myt3 in islets and to determine its significance in regulating islet function and survival. METHODS: Myt3 expression was determined in embryonic pancreas and adult islets by qPCR and immunohistochemistry. ChIP-seq, ChIP-qPCR and luciferase assays were used to evaluate regulation of Myt3 expression. Suppression of Myt3 was used to evaluate gene expression, insulin secretion and apoptosis in islets. RESULTS: We show that Myt3 is the most abundant MYT family member in adult islets and that it is expressed in all the major endocrine cell types in the pancreas after E18.5. We demonstrate that Myt3 expression is directly regulated by Foxa2, Pdx1, and Neurod1, which are critical to normal ß-cell development and function, and that Ngn3 induces Myt3 expression through alterations in the Myt3 promoter chromatin state. Further, we show that Myt3 expression is sensitive to both glucose and cytokine exposure. Of specific interest, suppressing Myt3 expression reduces insulin content and increases ß-cell apoptosis, at least in part, due to reduced Pdx1, Mafa, Il-6, Bcl-xl, c-Iap2 and Igfr1 levels, while over-expression of Myt3 protects islets from cytokine induced apoptosis. CONCLUSION/INTERPRETATION: We have identified Myt3 as a novel transcriptional regulator with a critical role in ß-cell survival. These data are an important step in clarifying the regulatory networks responsible for ß-cell survival, and point to Myt3 as a potential therapeutic target for improving functional ß-cell mass.


Subject(s)
Cell Survival/genetics , Gene Expression Regulation/physiology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Transcription Factors/metabolism , Zinc Fingers/genetics , Animals , Chromatin Immunoprecipitation , DNA Primers/genetics , Fluorescent Antibody Technique , Gene Expression Regulation/genetics , Immunohistochemistry , In Situ Hybridization , Luciferases , Mice , Polymerase Chain Reaction , RNA, Small Interfering/genetics , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...