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.
Diabetologia ; 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38958700

ABSTRACT

AIMS/HYPOTHESIS: Strategies to augment functional beta cell mass include directed differentiation of stem cells towards a beta cell fate, which requires extensive knowledge of transcriptional programs governing endocrine progenitor cell differentiation in vivo. We aimed to study the contributions of the Brahma-related gene-1 (BRG1) and Brahma (BRM) ATPase subunits of the SWI/SNF chromatin remodelling complex to endocrine cell development. METHODS: We generated mice with endocrine progenitor-specific Neurog3-Cre BRG1 removal in the presence of heterozygous (Brg1Δendo;Brm+/-) or homozygous (double knockout: DKOΔendo) BRM deficiency. Whole-body metabolic phenotyping, islet function characterisation, islet quantitative PCR and histological characterisation were performed on animals and tissues postnatally. To test the mechanistic actions of SWI/SNF in controlling gene expression during endocrine cell development, single-cell RNA-seq was performed on flow-sorted endocrine-committed cells from embryonic day 15.5 control and mutant embryos. RESULTS: Brg1Δendo;Brm+/- mice exhibit severe glucose intolerance, hyperglycaemia and hypoinsulinaemia, resulting, in part, from reduced islet number; diminished alpha, beta and delta cell mass; compromised islet insulin secretion; and altered islet gene expression programs, including reductions in MAFA and urocortin 3 (UCN3). DKOΔendo mice were not recovered at weaning; however, postnatal day 6 DKOΔendo mice were severely hyperglycaemic with reduced serum insulin levels and beta cell area. Single-cell RNA-seq of embryonic day 15.5 lineage-labelled cells revealed endocrine progenitor, alpha and beta cell populations from SWI/SNF mutants have reduced expression of Mafa, Gcg, Ins1 and Ins2, suggesting limited differentiation capacity. Reduced Neurog3 transcripts were discovered in DKOΔendo endocrine progenitor clusters, and the proliferative capacity of neurogenin 3 (NEUROG3)+ cells was reduced in Brg1Δendo;Brm+/- and DKOΔendo mutants. CONCLUSIONS/INTERPRETATION: Loss of BRG1 from developing endocrine progenitor cells has a severe postnatal impact on glucose homeostasis, and loss of both subunits impedes animal survival, with both groups exhibiting alterations in hormone transcripts embryonically. Taken together, these data highlight the critical role SWI/SNF plays in governing gene expression programs essential for endocrine cell development and expansion. DATA AVAILABILITY: Raw and processed data for scRNA-seq have been deposited into the NCBI Gene Expression Omnibus (GEO) database under the accession number GSE248369.

2.
Diabetes ; 72(6): 746-757, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36913741

ABSTRACT

The transcriptional activity of Pdx1 is modulated by a diverse array of coregulatory factors that govern chromatin accessibility, histone modifications, and nucleosome distribution. We previously identified the Chd4 subunit of the nucleosome remodeling and deacetylase complex as a Pdx1-interacting factor. To identify how loss of Chd4 impacts glucose homeostasis and gene expression programs in ß-cells in vivo, we generated an inducible ß-cell-specific Chd4 knockout mouse model. Removal of Chd4 from mature islet ß-cells rendered mutant animals glucose intolerant, in part due to defects in insulin secretion. We observed an increased ratio of immature-to-mature insulin granules in Chd4-deficient ß-cells that correlated with elevated levels of proinsulin both within isolated islets and from plasma following glucose stimulation in vivo. RNA sequencing and assay for transposase-accessible chromatin with sequencing showed that lineage-labeled Chd4-deficient ß-cells have alterations in chromatin accessibility and altered expression of genes critical for ß-cell function, including MafA, Slc2a2, Chga, and Chgb. Knockdown of CHD4 from a human ß-cell line revealed similar defects in insulin secretion and alterations in several ß-cell-enriched gene targets. These results illustrate how critical Chd4 activities are in controlling genes essential for maintaining ß-cell function. ARTICLE HIGHLIGHTS: Pdx1-Chd4 interactions were previously shown to be compromised in ß-cells from human donors with type 2 diabetes. ß-Cell-specific removal of Chd4 impairs insulin secretion and leads to glucose intolerance in mice. Expression of key ß-cell functional genes and chromatin accessibility are compromised in Chd4-deficient ß-cells. Chromatin remodeling activities enacted by Chd4 are essential for ß-cell function under normal physiological conditions.


Subject(s)
Chromatin , Diabetes Mellitus, Type 2 , Mice , Animals , Humans , Chromatin/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Diabetes Mellitus, Type 2/genetics , DNA Helicases/genetics , Mice, Knockout , Gene Expression , Glucose
3.
J Mol Endocrinol ; 69(2): 329-341, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35521759

ABSTRACT

Type 2 diabetes (T2D) is associated with loss of transcription factors (TFs) from a subset of failing ß-cells. Among these TFs is Pdx1, which controls the expression of numerous genes involved in maintaining ß-cell function and identity. Pdx1 activity is modulated by transcriptional coregulators and has recently been shown, through an unbiased screen, to interact with the Chd4 ATPase subunit of the nucleosome remodeling and deacetylase complex. Chd4 contributes to the maintenance of cellular identity and functional status of numerous different cell types. Here, we demonstrated that Pdx1 dynamically interacts with Chd4 under physiological and stimulatory conditions within islet ß-cells and established a fundamental role for Chd4 in regulating insulin secretion and modulating numerous Pdx1-bound genes in vitro, including the MafA TF, where we discovered Chd4 is bound to the MafA region 3 enhancer. Furthermore, we found that Pdx1:Chd4 interactions are significantly compromised in islet ß-cells under metabolically induced stress in vivo and in human donor tissues with T2D. Our findings establish a fundamental role for Chd4 in regulating insulin secretion and modulating Pdx1-bound genes in vitro, and disruption of Pdx1:Chd4 interactions coincides with ß-cell dysfunction associated with T2D.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Homeodomain Proteins/metabolism , Humans , Insulin-Secreting Cells/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...