Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Mol Metab ; 78: 101811, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37769990

ABSTRACT

OBJECTIVE: ASCL1, a pioneer transcription factor, is essential for neural cell differentiation and function. Previous studies have shown that Ascl1 expression is increased in pancreatic ß-cells lacking functional KATP channels or after feeding of a high fat diet (HFD) suggesting that it may contribute to the metabolic stress response of ß-cells. METHODS: We generated ß-cell-specific Ascl1 knockout mice (Ascl1ßKO) and assessed their glucose homeostasis, islet morphology and gene expression after feeding either a normal diet or HFD for 12 weeks, or in combination with a genetic disruption of Abcc8, an essential KATP channel component. RESULTS: Ascl1 expression is increased in response to both a HFD and membrane depolarization and requires CREB-dependent Ca2+ signaling. No differences in glucose homeostasis or islet morphology were observed in Ascl1ßKO mice fed a normal diet or in the absence of KATP channels. However, male Ascl1ßKO mice fed a HFD exhibited decreased blood glucose levels, improved glucose tolerance, and increased ß-cell proliferation. Bulk RNA-seq analysis of islets from Ascl1ßKO mice from three studied conditions showed alterations in genes associated with the secretory function. HFD-fed Ascl1ßKO mice showed the most extensive changes with increased expression of genes necessary for glucose sensing, insulin secretion and ß-cell proliferation, and a decrease in genes associated with ß-cell dysfunction, inflammation and dedifferentiation. HFD-fed Ascl1ßKO mice also displayed increased expression of parasympathetic neural markers and cholinergic receptors that was accompanied by increased insulin secretion in response to acetylcholine and an increase in islet innervation. CONCLUSIONS: Ascl1 expression is induced by stimuli that cause Ca2+-signaling to the nucleus and contributes in a multifactorial manner to the loss of ß-cell function by promoting the expression of genes associated with cellular dedifferentiation, attenuating ß-cells proliferation, suppressing acetylcholine sensitivity, and repressing parasympathetic innervation of islets. Thus, the removal of Ascl1 from ß-cells improves their function in response to metabolic stress.


Subject(s)
Acetylcholine , Basic Helix-Loop-Helix Transcription Factors , Insulin , Animals , Male , Mice , Adenosine Triphosphate/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Glucose , Insulin/metabolism , Insulin Secretion , Stress, Physiological
2.
JCI Insight ; 8(16)2023 08 22.
Article in English | MEDLINE | ID: mdl-37606041

ABSTRACT

Type 2 diabetes (T2D) is associated with compromised identity of insulin-producing pancreatic islet ß cells, characterized by inappropriate production of other islet cell-enriched hormones. Here, we examined how hormone misexpression was influenced by the MAFA and MAFB transcription factors, closely related proteins that maintain islet cell function. Mice specifically lacking MafA in ß cells demonstrated broad, population-wide changes in hormone gene expression with an overall gene signature closely resembling islet gastrin+ (Gast+) cells generated under conditions of chronic hyperglycemia and obesity. A human ß cell line deficient in MAFB, but not one lacking MAFA, also produced a GAST+ gene expression pattern. In addition, GAST was detected in human T2D ß cells with low levels of MAFB. Moreover, evidence is provided that human MAFB can directly repress GAST gene transcription. These results support a potentially novel, species-specific role for MafA and MAFB in maintaining adult mouse and human ß cell identity, respectively. Here, we discuss the possibility that induction of Gast/GAST and other non-ß cell hormones, by reduction in the levels of these transcription factors, represents a dysfunctional ß cell signature.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Islets of Langerhans , Adult , Humans , Animals , Mice , MafB Transcription Factor/genetics , Insulin
3.
PLoS Genet ; 19(5): e1010729, 2023 05.
Article in English | MEDLINE | ID: mdl-37155670

ABSTRACT

Repressive KRAB domain-containing zinc-finger proteins (KRAB-ZFPs) are abundant in mammalian genomes and contribute both to the silencing of transposable elements (TEs) and to the regulation of developmental stage- and cell type-specific gene expression. Here we describe studies of zinc finger protein 92 (Zfp92), an X-linked KRAB-ZFP that is highly expressed in pancreatic islets of adult mice, by analyzing global Zfp92 knockout (KO) mice. Physiological, transcriptomic and genome-wide chromatin binding studies indicate that the principal function of ZFP92 in mice is to bind to and suppress the activity of B1/Alu type of SINE elements and modulate the activity of surrounding genomic entities. Deletion of Zfp92 leads to changes in expression of select LINE and LTR retroelements and genes located in the vicinity of ZFP92-bound chromatin. The absence of Zfp92 leads to altered expression of specific genes in islets, adipose and muscle that result in modest sex-specific alterations in blood glucose homeostasis, body mass and fat accumulation. In islets, Zfp92 influences blood glucose concentration in postnatal mice via transcriptional effects on Mafb, whereas in adipose and muscle, it regulates Acacb, a rate-limiting enzyme in fatty acid metabolism. In the absence of Zfp92, a novel TE-Capn11 fusion transcript is overexpressed in islets and several other tissues due to de-repression of an IAPez TE adjacent to ZFP92-bound SINE elements in intron 3 of the Capn11 gene. Together, these studies show that ZFP92 functions both to repress specific TEs and to regulate the transcription of specific genes in discrete tissues.


Subject(s)
DNA Transposable Elements , Islets of Langerhans , Animals , Female , Male , Mice , Blood Glucose , Chromatin , Islets of Langerhans/metabolism , Mammals/genetics , Repressor Proteins/genetics , Retroelements/genetics , Zinc Fingers/genetics
4.
Stem Cells ; 41(6): 643-657, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37085274

ABSTRACT

During early embryogenesis, the transcription factor SOX17 contributes to hepato-pancreato-biliary system formation and vascular-hematopoietic emergence. To better understand Sox17 function in the developing endoderm and endothelium, we developed a dual-color temporal lineage-tracing strategy in mice combined with single-cell RNA sequencing to analyze 6934 cells from Sox17-expressing lineages at embryonic days 9.0-9.5. Our analyses showed 19 distinct cellular clusters combined from all 3 germ layers. Differential gene expression, trajectory and RNA-velocity analyses of endothelial cells revealed a heterogenous population of uncommitted and specialized endothelial subtypes, including 2 hemogenic populations that arise from different origins. Similarly, analyses of posterior foregut endoderm revealed subsets of hepatic, pancreatic, and biliary progenitors with overlapping developmental potency. Calculated gene-regulatory networks predict gene regulons that are dominated by cell type-specific transcription factors unique to each lineage. Vastly different Sox17 regulons found in endoderm versus endothelial cells support the differential interactions of SOX17 with other regulatory factors thereby enabling lineage-specific regulatory actions.


Subject(s)
Embryonic Development , Endothelial Cells , Gene Expression Regulation, Developmental , Gene Regulatory Networks , SOXF Transcription Factors , Animals , Mice , Cell Differentiation , Cell Lineage/genetics , Endoderm/metabolism , Endothelial Cells/metabolism , HMGB Proteins/genetics , HMGB Proteins/metabolism , Sequence Analysis, RNA , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Transcription Factors/metabolism , Embryonic Development/genetics
5.
iScience ; 26(2): 105938, 2023 Feb 17.
Article in English | MEDLINE | ID: mdl-36718359

ABSTRACT

Glucose stimulation induces the remodeling of microtubules, which potentiates insulin secretion in pancreatic ß-cells. CAMSAP2 binds to microtubule minus ends to stabilize microtubules in several cultured clonal cells. Here, we report that the knockdown of CAMSAP2 in primary ß-cells reduces total insulin content and attenuates GSIS without affecting the releasability of insulin vesicles. Surprisingly, CAMSAP2 knockdown does not change microtubule stability. Unlike in cultured insulinoma cells, CAMSAP2 in primary ß-cells predominantly localizes to the Golgi apparatus instead of microtubule minus ends. This novel localization is specific to primary ß- but not α-cells and is independent of microtubule binding. Consistent with its specific localization at the Golgi, CAMSAP2 promotes efficient Golgi-ER trafficking in primary ß-cells. Moreover, primary ß-cells and insulinoma cells likely express different CAMSAP2 isoforms. We propose that a novel CAMSAP2 isoform in primary ß-cells has a non-canonical function, which promotes Golgi-ER trafficking to support efficient production of insulin and secretion.

6.
Proc Natl Acad Sci U S A ; 119(49): e2213628119, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36442127

ABSTRACT

Single-nucleotide polymorphisms in the human juxtaposed with another zinc finger protein 1 (JAZF1) gene have repeatedly been associated with both type 2 diabetes (T2D) and height in multiple genome-wide association studies (GWAS); however, the mechanism by which JAZF1 causes these traits is not yet known. To investigate the possible functional role of JAZF1 in growth and glucose metabolism in vivo, we generated Jazf1 knockout (KO) mice and examined body composition and insulin sensitivity both in young and adult mice by using 1H-nuclear magnetic resonance and hyperinsulinemic-euglycemic clamp techniques. Plasma concentrations of insulin-like growth factor 1 (IGF-1) were reduced in both young and adult Jazf1 KO mice, and young Jazf1 KO mice were shorter in stature than age-matched wild-type mice. Young Jazf1 KO mice manifested reduced fat mass, whereas adult Jazf1 KO mice manifested increased fat mass and reductions in lean body mass associated with increased plasma growth hormone (GH) concentrations. Adult Jazf1 KO manifested muscle insulin resistance that was further exacerbated by high-fat diet feeding. Gene set enrichment analysis in Jazf1 KO liver identified the hepatocyte hepatic nuclear factor 4 alpha (HNF4α), which was decreased in Jazf1 KO liver and in JAZF1 knockdown cells. Moreover, GH-induced IGF-1 expression was inhibited by JAZF1 knockdown in human hepatocytes. Taken together these results demonstrate that reduction of JAZF1 leads to early growth retardation and late onset insulin resistance in vivo which may be mediated through alterations in the GH-IGF-1 axis and HNF4α.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Animals , Humans , Mice , Co-Repressor Proteins/genetics , Diabetes Mellitus, Type 2/genetics , DNA-Binding Proteins , Genome-Wide Association Study , Growth Disorders , Hepatocyte Nuclear Factor 4/genetics , Insulin Resistance/genetics , Insulin-Like Growth Factor I/genetics , Mice, Knockout
7.
iScience ; 25(9): 104905, 2022 Sep 16.
Article in English | MEDLINE | ID: mdl-36046192

ABSTRACT

Sox17 gene expression is essential for both endothelial and endodermal cell differentiation. To better understand the genetic basis for the expression of multiple Sox17 mRNA forms, we identified and performed CRISPR/Cas9 mutagenesis of two evolutionarily conserved promoter regions (CRs). The deletion of the upstream and endothelial cell-specific CR1 caused only a modest increase in lympho-vasculogenesis likely via reduced Notch signaling downstream of SOX17. In contrast, the deletion of the downstream CR2 region, which functions in both endothelial and endodermal cells, impairs both vascular and endodermal development causing death by embryonic day 12.5. Analyses of 3D chromatin looping, transcription factor binding, histone modification, and chromatin accessibility data at the Sox17 locus and surrounding region further support differential regulation of the two promoters during the development.

8.
Elife ; 102021 11 16.
Article in English | MEDLINE | ID: mdl-34783306

ABSTRACT

Heterogeneity of glucose-stimulated insulin secretion (GSIS) in pancreatic islets is physiologically important but poorly understood. Here, we utilize mouse islets to determine how microtubules (MTs) affect secretion toward the vascular extracellular matrix at single cell and subcellular levels. Our data indicate that MT stability in the ß-cell population is heterogenous, and that GSIS is suppressed in cells with highly stable MTs. Consistently, MT hyper-stabilization prevents, and MT depolymerization promotes the capacity of single ß-cell for GSIS. Analysis of spatiotemporal patterns of secretion events shows that MT depolymerization activates otherwise dormant ß-cells via initiation of secretion clusters (hot spots). MT depolymerization also enhances secretion from individual cells, introducing both additional clusters and scattered events. Interestingly, without MTs, the timing of clustered secretion is dysregulated, extending the first phase of GSIS and causing oversecretion. In contrast, glucose-induced Ca2+ influx was not affected by MT depolymerization yet required for secretion under these conditions, indicating that MT-dependent regulation of secretion hot spots acts in parallel with Ca2+ signaling. Our findings uncover a novel MT function in tuning insulin secretion hot spots, which leads to accurately measured and timed response to glucose stimuli and promotes functional ß-cell heterogeneity.


Subject(s)
Insulin Secretion , Insulin-Secreting Cells/metabolism , Microtubules/metabolism , Animals , Female , Insulin/metabolism , Male , Mice , Spatio-Temporal Analysis
9.
G3 (Bethesda) ; 11(11)2021 10 19.
Article in English | MEDLINE | ID: mdl-34534285

ABSTRACT

Insm1, Neurod1, and Pax6 are essential for the formation and function of pancreatic endocrine cells. Here, we report comparative immunohistochemical, transcriptomic, functional enrichment, and RNA splicing analyses of these genes using gene knock-out mice. Quantitative immunohistochemical analysis confirmed that elimination of each of these three factors variably impairs the proliferation, survival, and differentiation of endocrine cells. Transcriptomic analysis revealed that each factor contributes uniquely to the transcriptome although their effects were overlapping. Functional enrichment analysis revealed that genes downregulated by the elimination of Insm1, Neurod1, and Pax6 are commonly involved in mRNA metabolism, chromatin organization, secretion, and cell cycle regulation, and upregulated genes are associated with protein degradation, autophagy, and apoptotic process. Elimination of Insm1, Neurod1, and Pax6 impaired expression of many RNA-binding proteins thereby altering RNA splicing events, including for Syt14 and Snap25, two genes required for insulin secretion. All three factors are necessary for normal splicing of Syt14, and both Insm1 and Pax6 are necessary for the processing of Snap25. Collectively, these data provide new insights into how Insm1, Neurod1, and Pax6 contribute to the formation of functional pancreatic endocrine cells.


Subject(s)
Endocrine Cells , Transcription Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Endocrine Cells/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Mice , PAX6 Transcription Factor/genetics , RNA , RNA Splicing , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics
10.
Development ; 148(6)2021 03 21.
Article in English | MEDLINE | ID: mdl-33653874

ABSTRACT

To gain a deeper understanding of pancreatic ß-cell development, we used iterative weighted gene correlation network analysis to calculate a gene co-expression network (GCN) from 11 temporally and genetically defined murine cell populations. The GCN, which contained 91 distinct modules, was then used to gain three new biological insights. First, we found that the clustered protocadherin genes are differentially expressed during pancreas development. Pcdhγ genes are preferentially expressed in pancreatic endoderm, Pcdhß genes in nascent islets, and Pcdhα genes in mature ß-cells. Second, after extracting sub-networks of transcriptional regulators for each developmental stage, we identified 81 zinc finger protein (ZFP) genes that are preferentially expressed during endocrine specification and ß-cell maturation. Third, we used the GCN to select three ZFPs for further analysis by CRISPR mutagenesis of mice. Zfp800 null mice exhibited early postnatal lethality, and at E18.5 their pancreata exhibited a reduced number of pancreatic endocrine cells, alterations in exocrine cell morphology, and marked changes in expression of genes involved in protein translation, hormone secretion and developmental pathways in the pancreas. Together, our results suggest that developmentally oriented GCNs have utility for gaining new insights into gene regulation during organogenesis.


Subject(s)
Cell Differentiation/genetics , Homeodomain Proteins/genetics , Organogenesis/genetics , Pancreas/growth & development , Animals , Cadherins/genetics , Cell Lineage/genetics , Gene Expression Regulation, Developmental/genetics , Insulin/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mice , Pancreas/metabolism
11.
Diabetes ; 69(9): 1936-1947, 2020 09.
Article in English | MEDLINE | ID: mdl-32540877

ABSTRACT

The microtubule cytoskeleton of pancreatic islet ß-cells regulates glucose-stimulated insulin secretion (GSIS). We have reported that the microtubule-mediated movement of insulin vesicles away from the plasma membrane limits insulin secretion. High glucose-induced remodeling of microtubule network facilitates robust GSIS. This remodeling involves disassembly of old microtubules and nucleation of new microtubules. Here, we examine the mechanisms whereby glucose stimulation decreases microtubule lifetimes in ß-cells. Using real-time imaging of photoconverted microtubules, we demonstrate that high levels of glucose induce rapid microtubule disassembly preferentially in the periphery of individual ß-cells, and this process is mediated by the phosphorylation of microtubule-associated protein tau. Specifically, high glucose induces tau hyper-phosphorylation via glucose-responsive kinases GSK3, PKA, PKC, and CDK5. This causes dissociation of tau from and subsequent destabilization of microtubules. Consequently, tau knockdown in mouse islet ß-cells facilitates microtubule turnover, causing increased basal insulin secretion, depleting insulin vesicles from the cytoplasm, and impairing GSIS. More importantly, tau knockdown uncouples microtubule destabilization from glucose stimulation. These findings suggest that tau suppresses peripheral microtubules turning over to restrict insulin oversecretion in basal conditions and preserve the insulin pool that can be released following stimulation; high glucose promotes tau phosphorylation to enhance microtubule disassembly to acutely enhance GSIS.


Subject(s)
Glucose/pharmacology , Insulin Secretion/drug effects , Insulin-Secreting Cells/drug effects , Microtubules/drug effects , tau Proteins/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclin-Dependent Kinase 5/metabolism , Glycogen Synthase Kinase 3/metabolism , Insulin-Secreting Cells/metabolism , Mice , Microtubules/metabolism , Phosphorylation/drug effects , Protein Kinase C
12.
Diabetes ; 69(7): 1476-1491, 2020 07.
Article in English | MEDLINE | ID: mdl-32332159

ABSTRACT

A sustained increase in intracellular Ca2+ concentration (referred to hereafter as excitotoxicity), brought on by chronic metabolic stress, may contribute to pancreatic ß-cell failure. To determine the additive effects of excitotoxicity and overnutrition on ß-cell function and gene expression, we analyzed the impact of a high-fat diet (HFD) on Abcc8 knockout mice. Excitotoxicity caused ß-cells to be more susceptible to HFD-induced impairment of glucose homeostasis, and these effects were mitigated by verapamil, a Ca2+ channel blocker. Excitotoxicity, overnutrition, and the combination of both stresses caused similar but distinct alterations in the ß-cell transcriptome, including additive increases in genes associated with mitochondrial energy metabolism, fatty acid ß-oxidation, and mitochondrial biogenesis and their key regulator Ppargc1a Overnutrition worsened excitotoxicity-induced mitochondrial dysfunction, increasing metabolic inflexibility and mitochondrial damage. In addition, excitotoxicity and overnutrition, individually and together, impaired both ß-cell function and identity by reducing expression of genes important for insulin secretion, cell polarity, cell junction, cilia, cytoskeleton, vesicular trafficking, and regulation of ß-cell epigenetic and transcriptional program. Sex had an impact on all ß-cell responses, with male animals exhibiting greater metabolic stress-induced impairments than females. Together, these findings indicate that a sustained increase in intracellular Ca2+, by altering mitochondrial function and impairing ß-cell identity, augments overnutrition-induced ß-cell failure.


Subject(s)
Calcium/metabolism , Insulin-Secreting Cells/metabolism , Overnutrition/metabolism , Animals , Cells, Cultured , Diet, High-Fat , Female , Gene Expression Regulation , Glucose/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/physiology , Sex Characteristics , Transcriptome
13.
Nat Genet ; 52(4): 418-427, 2020 04.
Article in English | MEDLINE | ID: mdl-32203463

ABSTRACT

Gene network transitions in embryos and other fate-changing contexts involve combinations of transcription factors. A subset of fate-changing transcription factors act as pioneers; they scan and target nucleosomal DNA and initiate cooperative events that can open the local chromatin. However, a gap has remained in understanding how molecular interactions with the nucleosome contribute to the chromatin-opening phenomenon. Here we identified a short α-helical region, conserved among FOXA pioneer factors, that interacts with core histones and contributes to chromatin opening in vitro. The same domain is involved in chromatin opening in early mouse embryos for normal development. Thus, local opening of chromatin by interactions between pioneer factors and core histones promotes genetic programming.


Subject(s)
Gene Regulatory Networks/genetics , Histones/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Cell Line , Chromatin/genetics , DNA/genetics , Female , Gene Expression Regulation, Developmental/genetics , Humans , Mice , Mice, Inbred C57BL , Nucleosomes/genetics , Transcription, Genetic/genetics
14.
Diabetes ; 68(9): 1806-1818, 2019 09.
Article in English | MEDLINE | ID: mdl-31201281

ABSTRACT

Transcription factors positively and/or negatively impact gene expression by recruiting coregulatory factors, which interact through protein-protein binding. Here we demonstrate that mouse pancreas size and islet ß-cell function are controlled by the ATP-dependent Swi/Snf chromatin remodeling coregulatory complex that physically associates with Pdx1, a diabetes-linked transcription factor essential to pancreatic morphogenesis and adult islet cell function and maintenance. Early embryonic deletion of just the Swi/Snf Brg1 ATPase subunit reduced multipotent pancreatic progenitor cell proliferation and resulted in pancreas hypoplasia. In contrast, removal of both Swi/Snf ATPase subunits, Brg1 and Brm, was necessary to compromise adult islet ß-cell activity, which included whole-animal glucose intolerance, hyperglycemia, and impaired insulin secretion. Notably, lineage-tracing analysis revealed Swi/Snf-deficient ß-cells lost the ability to produce the mRNAs for Ins and other key metabolic genes without effecting the expression of many essential islet-enriched transcription factors. Swi/Snf was necessary for Pdx1 to bind to the Ins gene enhancer, demonstrating the importance of this association in mediating chromatin accessibility. These results illustrate how fundamental the Pdx1:Swi/Snf coregulator complex is in the pancreas, and we discuss how disrupting their association could influence type 1 and type 2 diabetes susceptibility.


Subject(s)
Cell Proliferation/physiology , Chromatin Assembly and Disassembly/physiology , DNA Helicases/metabolism , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/metabolism , Nuclear Proteins/metabolism , Pancreas/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , DNA Helicases/genetics , Gene Expression Regulation , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Homeodomain Proteins/genetics , Insulin/blood , Insulin-Secreting Cells/cytology , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Pancreas/cytology , Trans-Activators/genetics , Transcription Factors/genetics
15.
Am J Physiol Endocrinol Metab ; 316(2): E196-E209, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30532991

ABSTRACT

Fluorescent protein reporter genes are widely used to identify and sort murine pancreatic ß-cells. In this study, we compared use of the MIP-GFP transgene, which exhibits aberrant expression of human growth hormone (hGH), with a newly derived Ins2Apple allele that lacks hGH expression on the expression of sex-specific genes. ß-Cells from MIP-GFP transgenic mice exhibit changes in the expression of 7,733 genes, or greater than half of their transcriptome, compared with ß-cells from Ins2Apple/+ mice. To determine how these differences might affect a typical differential gene expression study, we analyzed the effect of sex on gene expression using both reporter lines. Six hundred fifty-seven differentially expressed genes were identified between male and female ß-cells containing the Ins2Apple allele. Female ß-cells exhibit higher expression of Xist, Tmed9, Arpc3, Eml2, and several islet-enriched transcription factors, including Nkx2-2 and Hnf4a, whereas male ß-cells exhibited a generally higher expression of genes involved in cell cycle regulation. In marked contrast, the same male vs. female comparison of ß-cells containing the MIP-GFP transgene revealed only 115 differentially expressed genes, and comparison of the 2 lists of differentially expressed genes revealed only 17 that were common to both analyses. These results indicate that 1) male and female ß-cells differ in their expression of key transcription factors and cell cycle regulators and 2) the MIP-GFP transgene may attenuate sex-specific differences that distinguish male and female ß-cells, thereby impairing the identification of sex-specific variations.


Subject(s)
Green Fluorescent Proteins/genetics , Human Growth Hormone/genetics , Insulin-Secreting Cells/metabolism , Insulin/genetics , Animals , Female , Gene Expression , Genes, Reporter/genetics , Green Fluorescent Proteins/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Human Growth Hormone/metabolism , Humans , Male , Mice , Mice, Transgenic , Nuclear Proteins , Promoter Regions, Genetic , RNA, Messenger/metabolism , Sex Factors , Transcription Factors , Transgenes
16.
Nat Cell Biol ; 20(11): 1267-1277, 2018 11.
Article in English | MEDLINE | ID: mdl-30361701

ABSTRACT

The mechanisms that restrict regeneration and maintain cell identity following injury are poorly characterized in higher vertebrates. Following ß-cell loss, 1-2% of the glucagon-producing α-cells spontaneously engage in insulin production in mice. Here we explore the mechanisms inhibiting α-cell plasticity. We show that adaptive α-cell identity changes are constrained by intra-islet insulin- and Smoothened-mediated signalling, among others. The combination of ß-cell loss or insulin-signalling inhibition, with Smoothened inactivation in α- or δ-cells, stimulates insulin production in more α-cells. These findings suggest that the removal of constitutive 'brake signals' is crucial to neutralize the refractoriness to adaptive cell-fate changes. It appears that the maintenance of cell identity is an active process mediated by repressive signals, which are released by neighbouring cells and curb an intrinsic trend of differentiated cells to change.


Subject(s)
Insulin/metabolism , Islets of Langerhans/metabolism , Signal Transduction , Smoothened Receptor/metabolism , Animals , Cell Differentiation , Cell Plasticity , Cell Proliferation , Female , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Mice, Transgenic , Smoothened Receptor/genetics
17.
Cell Stem Cell ; 22(1): 12-13, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29304337

ABSTRACT

Beta cell replacement strategies hold promise for permanently treating type 1 diabetes. In Cell Stem Cell, Xiao et al. (2018) restore pancreatic beta cell mass and normalize blood glucose in diabetic mice by reprogramming pancreatic alpha to beta cells using Pdx1- and Mafa-expressing adeno-associated virus infused into the pancreatic duct.


Subject(s)
Cellular Reprogramming , Insulin-Secreting Cells , Animals , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Homeodomain Proteins/genetics , Insulin/genetics , Mice , Pancreatic Ducts
18.
Diabetes ; 66(8): 2175-2187, 2017 08.
Article in English | MEDLINE | ID: mdl-28550109

ABSTRACT

We used mice lacking Abcc8, a key component of the ß-cell KATP-channel, to analyze the effects of a sustained elevation in the intracellular Ca2+ concentration ([Ca2+]i) on ß-cell identity and gene expression. Lineage tracing analysis revealed the conversion of ß-cells lacking Abcc8 into pancreatic polypeptide cells but not to α- or δ-cells. RNA-sequencing analysis of FACS-purified Abcc8-/- ß-cells confirmed an increase in Ppy gene expression and revealed altered expression of more than 4,200 genes, many of which are involved in Ca2+ signaling, the maintenance of ß-cell identity, and cell adhesion. The expression of S100a6 and S100a4, two highly upregulated genes, is closely correlated with membrane depolarization, suggesting their use as markers for an increase in [Ca2+]i Moreover, a bioinformatics analysis predicts that many of the dysregulated genes are regulated by common transcription factors, one of which, Ascl1, was confirmed to be directly controlled by Ca2+ influx in ß-cells. Interestingly, among the upregulated genes is Aldh1a3, a putative marker of ß-cell dedifferentiation, and other genes associated with ß-cell failure. Taken together, our results suggest that chronically elevated ß-cell [Ca2+]i in Abcc8-/- islets contributes to the alteration of ß-cell identity, islet cell numbers and morphology, and gene expression by disrupting a network of Ca2+-regulated genes.


Subject(s)
Calcium Signaling/genetics , Cell Polarity , Gene Expression Regulation/genetics , Gene Expression/genetics , Insulin-Secreting Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/physiology , Calcium/metabolism , Cell Adhesion/genetics , Cell Cycle Proteins/metabolism , Cell Lineage/genetics , Insulin-Secreting Cells/cytology , KATP Channels/genetics , Mice , Pancreatic Polypeptide-Secreting Cells/physiology , S100 Calcium Binding Protein A6 , S100 Calcium-Binding Protein A4/metabolism , S100 Proteins/metabolism , Sulfonylurea Receptors/deficiency
19.
Cell Rep ; 17(8): 2028-2041, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27851966

ABSTRACT

Using a transgenic mouse model to express MafA, Pdx1, and Neurog3 (3TF) in a pancreatic acinar cell- and doxycycline-dependent manner, we discovered that the outcome of transcription factor-mediated acinar to ß-like cellular reprogramming is dependent on both the magnitude of 3TF expression and on reprogramming-induced inflammation. Overly robust 3TF expression causes acinar cell necrosis, resulting in marked inflammation and acinar-to-ductal metaplasia. Generation of new ß-like cells requires limiting reprogramming-induced inflammation, either by reducing 3TF expression or by eliminating macrophages. The new ß-like cells were able to reverse streptozotocin-induced diabetes 6 days after inducing 3TF expression but failed to sustain their function after removal of the reprogramming factors.


Subject(s)
Acinar Cells/pathology , Cellular Reprogramming , Inflammation/pathology , Insulin-Secreting Cells/pathology , Pancreas/pathology , Acinar Cells/drug effects , Adenoviridae/metabolism , Alleles , Animals , Cellular Reprogramming/drug effects , Diabetes Mellitus, Experimental/pathology , Doxycycline/pharmacology , Gene Expression Profiling , Homeodomain Proteins/metabolism , Immunity , Insulin-Secreting Cells/drug effects , Macrophages/drug effects , Macrophages/pathology , Metaplasia , Mice, Transgenic , Organ Size/drug effects , Pancreatic Ducts/pathology , Reproducibility of Results , Transcription Factors/metabolism , Transgenes
20.
Development ; 143(24): 4595-4607, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27864380

ABSTRACT

SET domain-containing proteins play a vital role in regulating gene expression during development through modifications in chromatin structure. Here we show that SET domain-containing 5 (Setd5) is divergently transcribed with Gt(ROSA26)Sor, is necessary for mammalian development, and interacts with the PAF1 co-transcriptional complex and other proteins. Setd5-deficient mouse embryos exhibit severe defects in neural tube formation, somitogenesis and cardiac development, have aberrant vasculogenesis in embryos, yolk sacs and placentas, and die between embryonic day 10.5 and 11.5. Setd5-deficient embryonic stem cells have impaired cellular proliferation, increased apoptosis, defective cell cycle progression, a diminished ability to differentiate into cardiomyocytes and greatly perturbed gene expression. SETD5 co-immunoprecipitates with multiple components of the PAF1 and histone deacetylase-containing NCoR complexes and is not solely required for major histone lysine methylation marks. In the absence of Setd5, histone acetylation is increased at transcription start sites and near downstream regions. These findings suggest that SETD5 functions in a manner similar to yeast Set3p and Drosophila UpSET, and that it is essential for regulating histone acetylation during gene transcription.


Subject(s)
Chromatin/genetics , Embryonic Development/genetics , Gene Expression Regulation/genetics , Histones/metabolism , Methyltransferases/genetics , Acetylation , Animals , Apoptosis/genetics , Carrier Proteins/metabolism , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Embryonic Stem Cells/cytology , Heart Defects, Congenital/genetics , Methyltransferases/metabolism , Mice , Mice, Knockout , Myocytes, Cardiac/cytology , Neural Tube/growth & development , Promoter Regions, Genetic/genetics , RNA, Untranslated/genetics , Transcription, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...