Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
FEBS J ; 291(3): 566-583, 2024 02.
Article in English | MEDLINE | ID: mdl-37985375

ABSTRACT

Butyrate, a gut microbial metabolite, has beneficial effects on glucose homeostasis and has become an attractive drug candidate for type 2 diabetes (T2D). Recently, we showed that butyrate protects pancreatic beta cells against cytokine-induced dysfunction. In this study, we explored the underlying mechanisms of butyrate action. Pancreatic mouse islets were exposed to a non-cytotoxic concentration of interleukin-1ß (IL-1ß) for 10 days to mimic low-grade inflammation in T2D. Similar to the effect of butyrate, an isoform-selective histone deacetylase 3 (HDAC3) inhibitor normalized IL-1ß-reduced glucose-stimulated insulin secretion and insulin content. In contrast, free fatty acid receptor 2 and 3 (FFAR2/3) agonists failed to normalize IL-1ß-induced beta cell dysfunction. Furthermore, butyrate inhibited HDAC activity and increased the acetylation of histone H3 and H4 by 3- and 10-fold, respectively. Genome-wide analysis of histone H3 lysine 27 acetylation (H3K27ac) revealed that butyrate mainly increased H3K27ac at promoter regions (74%), while H3K27ac peaks regulated by IL-1ß were more equally distributed at promoters (38%), introns (23%) and intergenic regions (23%). Gene ontology analysis showed that butyrate increased IL-1ß-reduced H3K27ac levels near several genes related to hormone secretion and reduced IL-1ß-increased H3K27ac levels near genes associated with inflammatory responses. Butyrate alone increased H3K27ac near many genes related to MAPK signaling, hormone secretion, and differentiation, and decreased H3K27ac at genes involved in cell replication. Together, these results suggest that butyrate prevents IL-1ß-induced pancreatic islet dysfunction by inhibition of HDACs resulting in changes in H3K27ac levels at genes relevant for beta cell function and inflammatory responses.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Mice , Animals , Histone Deacetylase Inhibitors/pharmacology , Interleukin-1beta/metabolism , Histones/genetics , Histones/metabolism , Insulin-Secreting Cells/metabolism , Butyrates/pharmacology , Butyrates/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Hormones/metabolism
2.
Cell Death Dis ; 14(7): 399, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37407581

ABSTRACT

Insufficient insulin secretion is a hallmark of type 2 diabetes and has been attributed to beta cell identity loss characterized by decreased expression of several key beta cell genes. The pro-inflammatory factor BMP-2 is upregulated in islets of Langerhans from individuals with diabetes and acts as an inhibitor of beta cell function and proliferation. Exposure to BMP-2 induces expression of Id1-4, Hes-1, and Hey-1 which are transcriptional regulators associated with loss of differentiation. The aim of this study was to investigate the mechanism by which BMP-2 induces beta cell dysfunction and loss of cell maturity. Mouse islets exposed to BMP-2 for 10 days showed impaired glucose-stimulated insulin secretion and beta cell proliferation. BMP-2-induced beta cell dysfunction was associated with decreased expression of cell maturity and proliferation markers specific to the beta cell such as Ins1, Ucn3, and Ki67 and increased expression of Id1-4, Hes-1, and Hey-1. The top 30 most regulated proteins significantly correlated with corresponding mRNA expression. BMP-2-induced gene expression changes were associated with a predominant reduction in acetylation of H3K27 and a decrease in NeuroD1 chromatin binding activity. These results show that BMP-2 induces loss of beta cell maturity and suggest that remodeling of H3K27ac and decreased NeuroD1 DNA binding activity participate in the effect of BMP-2 on beta cell dysfunction.


Subject(s)
Diabetes Mellitus, Type 2 , Histone Code , Animals , Mice , Bone Morphogenetic Protein 2/metabolism , Chromatin , Diabetes Mellitus, Type 2/metabolism , Protein Processing, Post-Translational , Signal Transduction
3.
Int J Mol Sci ; 24(5)2023 Feb 25.
Article in English | MEDLINE | ID: mdl-36901964

ABSTRACT

Butyrate produced by the gut microbiota has beneficial effects on metabolism and inflammation. Butyrate-producing bacteria are supported by diets with a high fiber content, such as high-amylose maize starch (HAMS). We investigated the effects of HAMS- and butyrylated HAMS (HAMSB)-supplemented diets on glucose metabolism and inflammation in diabetic db/db mice. Mice fed HAMSB had 8-fold higher fecal butyrate concentration compared to control diet-fed mice. Weekly analysis of fasting blood glucose showed a significant reduction in HAMSB-fed mice when the area under the curve for all five weeks was analyzed. Following treatment, fasting glucose and insulin analysis showed increased homeostatic model assessment (HOMA) insulin sensitivity in the HAMSB-fed mice. Glucose-stimulated insulin release from isolated islets did not differ between the groups, while insulin content was increased by 36% in islets of the HAMSB-fed mice. Expression of insulin 2 was also significantly increased in islets of the HAMSB-fed mice, while no difference in expression of insulin 1, pancreatic and duodenal homeobox 1, MAF bZIP transcription factor A and urocortin 3 between the groups was observed. Hepatic triglycerides in the livers of the HAMSB-fed mice were significantly reduced. Finally, mRNA markers of inflammation in liver and adipose tissue were reduced in mice fed HAMSB. These findings suggest that HAMSB-supplemented diet improves glucose metabolism in the db/db mice, and reduces inflammation in insulin-sensitive tissues.


Subject(s)
Butyrates , Starch , Rats , Mice , Animals , Rats, Sprague-Dawley , Amylose/metabolism , Inflammation , Liver/metabolism , Mice, Inbred Strains , Insulin , Homeostasis , Glucose , Mice, Inbred C57BL , Blood Glucose/metabolism
4.
J Biol Chem ; 298(9): 102312, 2022 09.
Article in English | MEDLINE | ID: mdl-35921894

ABSTRACT

Cytokine-induced beta cell dysfunction is a hallmark of type 2 diabetes (T2D). Chronic exposure of beta cells to inflammatory cytokines affects gene expression and impairs insulin secretion. Thus, identification of anti-inflammatory factors that preserve beta cell function represents an opportunity to prevent or treat T2D. Butyrate is a gut microbial metabolite with anti-inflammatory properties for which we recently showed a role in preventing interleukin-1ß (IL-1ß)-induced beta cell dysfunction, but how prevention is accomplished is unclear. Here, we investigated the mechanisms by which butyrate exerts anti-inflammatory activity in beta cells. We exposed mouse islets and INS-1E cells to a low dose of IL-1ß and/or butyrate and measured expression of inflammatory genes and nitric oxide (NO) production. Additionally, we explored the molecular mechanisms underlying butyrate activity by dissecting the activation of the nuclear factor-κB (NF-κB) pathway. We found that butyrate suppressed IL-1ß-induced expression of inflammatory genes, such as Nos2, Cxcl1, and Ptgs2, and reduced NO production. Butyrate did not inhibit IκBα degradation nor NF-κB p65 nuclear translocation. Furthermore, butyrate did not affect binding of NF-κB p65 to target sequences in synthetic DNA but inhibited NF-κB p65 binding and RNA polymerase II recruitment to inflammatory gene promoters in the context of native DNA. We found this was concurrent with increased acetylation of NF-κB p65 and histone H4, suggesting butyrate affects NF-κB activity via inhibition of histone deacetylases. Together, our results show butyrate inhibits IL-1ß-induced inflammatory gene expression and NO production through suppression of NF-κB activation and thereby possibly preserves beta cell function.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Butyrates , Diabetes Mellitus, Type 2 , Histone Deacetylase Inhibitors , Inflammation , Insulin-Secreting Cells , Interleukin-1beta , NF-kappa B , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Butyrates/pharmacology , Cyclooxygenase 2/metabolism , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/pathology , Gene Expression Regulation , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Histones/metabolism , Inflammation/genetics , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Mice , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , RNA Polymerase II/metabolism
5.
Int J Mol Sci ; 22(19)2021 Sep 27.
Article in English | MEDLINE | ID: mdl-34638768

ABSTRACT

Pancreatic beta cell dysfunction caused by metabolic and inflammatory stress contributes to the development of type 2 diabetes (T2D). Butyrate, produced by the gut microbiota, has shown beneficial effects on glucose metabolism in animals and humans and may directly affect beta cell function, but the mechanisms are poorly described. The aim of this study was to investigate the effect of butyrate on cytokine-induced beta cell dysfunction in vitro. Mouse islets, rat INS-1E, and human EndoC-ßH1 beta cells were exposed long-term to non-cytotoxic concentrations of cytokines and/or butyrate to resemble the slow onset of inflammation in T2D. Beta cell function was assessed by glucose-stimulated insulin secretion (GSIS), gene expression by qPCR and RNA-sequencing, and proliferation by incorporation of EdU into newly synthesized DNA. Butyrate protected beta cells from cytokine-induced impairment of GSIS and insulin content in the three beta cell models. Beta cell proliferation was reduced by both cytokines and butyrate. Expressions of the beta cell specific genes Ins, MafA, and Ucn3 reduced by the cytokine IL-1ß were not affected by butyrate. In contrast, butyrate upregulated the expression of secretion/transport-related genes and downregulated inflammatory genes induced by IL-1ß in mouse islets. In summary, butyrate prevents pro-inflammatory cytokine-induced beta cell dysfunction.


Subject(s)
Butyrates/pharmacology , Diabetes Mellitus, Type 2/immunology , Gene Expression Regulation/drug effects , Insulin-Secreting Cells/immunology , Interleukin-1beta/immunology , Animals , Cell Line , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Humans , Insulin-Secreting Cells/pathology , Male , Mice , Rats
6.
Mol Cell Endocrinol ; 496: 110524, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31362031

ABSTRACT

Decreased insulin secretory capacity in Type 2 diabetes mellitus is associated with beta-cell dedifferentiation and inflammation. We hypothesize that prolonged exposure of beta-cells to low concentrations of IL-1ß induce beta-cell dedifferentiation characterized by impaired glucose-stimulated insulin secretion, reduced expression of key beta-cell genes and changes in histone modifications at gene loci known to affect beta-cell function. Ten days exposure to IL-1ß at non-cytotoxic concentrations reduced insulin secretion and beta-cell proliferation and decreased expression of key beta-cell identity genes, including MafA and Ucn3 and decreased H3K27ac at the gene loci, suggesting that inflammatory cytokines directly affects the epigenome. Following removal of IL-1ß, beta-cell function was normalized and mRNA expression of beta-cell identity genes, such as insulin and Ucn3 returned to pre-stimulation levels. Our findings indicate that prolonged exposure to low concentrations of IL-1ß induces epigenetic changes associated with loss of beta-cell identity as observed in Type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation , Histones/metabolism , Insulin-Secreting Cells/metabolism , Interleukin-1beta/metabolism , Protein Processing, Post-Translational , Animals , Diabetes Mellitus, Type 2/pathology , Epigenesis, Genetic , Insulin-Secreting Cells/pathology , Maf Transcription Factors, Large/biosynthesis , Male , Mice , Urocortins/biosynthesis
7.
Mol Cell Proteomics ; 17(1): 95-110, 2018 01.
Article in English | MEDLINE | ID: mdl-29113996

ABSTRACT

Normal pancreatic islet ß-cells (PBCs) abundantly secrete insulin in response to elevated blood glucose levels, in order to maintain an adequate control of energy balance and glucose homeostasis. However, the molecular mechanisms underlying the insulin secretion are unclear. Improving our understanding of glucose-stimulated insulin secretion (GSIS) mechanisms under normal conditions is a prerequisite for developing better interventions against diabetes. Here, we aimed at identifying novel signaling pathways involved in the initial release of insulin from PBCs after glucose stimulation using quantitative strategies for the assessment of phosphorylated proteins and sialylated N-linked (SA) glycoproteins.Islets of Langerhans derived from newborn rats with a subsequent 9-10 days of maturation in vitro were stimulated with 20 mm glucose for 0 min (control), 5 min, 10 min, and 15 min. The isolated islets were subjected to time-resolved quantitative phosphoproteomics and sialiomics using iTRAQ-labeling combined with enrichment of phosphorylated peptides and formerly SA glycopeptides and high-accuracy LC-MS/MS. Using bioinformatics we analyzed the functional signaling pathways during GSIS, including well-known insulin secretion pathways. Furthermore, we identified six novel activated signaling pathways (e.g. agrin interactions and prolactin signaling) at 15 min GSIS, which may increase our understanding of the molecular mechanism underlying GSIS. Moreover, we validated some of the regulated phosphosites by parallel reaction monitoring, which resulted in the validation of eleven new phosphosites significantly regulated on GSIS. Besides protein phosphorylation, alteration in SA glycosylation was observed on several surface proteins on brief GSIS. Interestingly, proteins important for cell-cell interaction, cell movement, cell-ECM interaction and Focal Adhesion (e.g. integrins, semaphorins, and plexins) were found regulated at the level of sialylation, but not in protein expression. Collectively, we believe that this comprehensive Proteomics and PTMomics survey of signaling pathways taking place during brief GSIS of primary PBCs is contributing to understanding the complex signaling underlying GSIS.


Subject(s)
Glucose/pharmacology , Insulin Secretion/physiology , Insulin-Secreting Cells/drug effects , Animals , Animals, Newborn , Insulin-Secreting Cells/metabolism , Protein Processing, Post-Translational , Proteomics , Rats, Wistar , Signal Transduction
8.
Eur J Pharmacol ; 815: 181-189, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28928089

ABSTRACT

GLP-1-gastrin dual agonist ZP3022 has been shown to increase ß-cell mass with a concomitant improvement of glycemic control in diabetic mice and rats. Here we tested the in-vitro effects of ZP3022 on ß-cell proliferation, islet apoptosis and glucose-stimulated insulin secretion (GSIS) in rat islets of Langerhans. Moreover, gene expression profiling in whole pancreas from Zucker Diabetic Fatty (ZDF) rats was performed to characterize genes differently regulated by short-term treatment with ZP3022. Treatments with exendin-4, gastrin-17 alone or in combination were included in the studies. ZP3022 promoted ß-cell proliferation, protected from palmitate-, but not from cytokine-induced apoptosis, and induced an increase in GSIS, demonstrating a glucose dependent insulinotropic action of ZP3022 on ß-cells. The combination treatment with exendin-4 and gastrin-17 showed comparable effects on proliferation, apoptosis, and GSIS as did ZP3022. Microarray analysis revealed that ZP3022 exerted specific effects on pancreatic gene expression not observed when treating ZDF rats with either exendin-4 alone or in combination with gastrin-17. In particular MAPK signaling pathway was observed among the highest affected pathways; while also pathways related to insulin signaling and secretion were regulated by ZP3022. Moreover, rats treated with ZP3022 had a higher expression of genes encoding for the specific ß-cell/endocrine cell markers, such as islet amyloid polypeptide (IAPP), protein convertase 1/3 and -2 (PC 1/3 and-2), as well as transmembrane protein 27(TMEM27) compared to vehicle treated rats. We conclude that ZP3022 may have therapeutic potential in the prevention/delay of ß cell dysfunction.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Peptides/pharmacology , Amino Acid Sequence , Animals , Apoptosis/drug effects , Blood Glucose/metabolism , Body Weight/drug effects , Cell Proliferation/drug effects , Cytokines/pharmacology , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Palmitic Acid/pharmacology , Peptides/chemistry , Peptides/therapeutic use , Rats , Rats, Wistar
9.
Mol Cell Endocrinol ; 448: 108-121, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28390953

ABSTRACT

Pannexins (Panx's) are membrane proteins involved in a variety of biological processes, including cell death signaling and immune functions. The role and functions of Panx's in pancreatic ß-cells remain to be clarified. Here, we show Panx1 and Panx2 expression in isolated islets, primary ß-cells, and ß-cell lines. The expression of Panx2, but not Panx1, was downregulated by interleukin-1ß (IL-1ß) plus interferon-γ (IFNγ), two pro-inflammatory cytokines suggested to contribute to ß-cell demise in type 1 diabetes (T1D). siRNA-mediated knockdown (KD) of Panx2 aggravated cytokine-induced apoptosis in rat INS-1E cells and primary rat ß-cells, suggesting anti-apoptotic properties of Panx2. An anti-apoptotic function of Panx2 was confirmed in isolated islets from Panx2-/- mice and in human EndoC-ßH1 cells. Panx2 KD was associated with increased cytokine-induced activation of STAT3 and higher expression of inducible nitric oxide synthase (iNOS). Glucose-stimulated insulin release was impaired in Panx2-/- islets, and Panx2-/- mice subjected to multiple low-dose Streptozotocin (MLDS) treatment, a model of T1D, developed more severe diabetes compared to wild type mice. These data suggest that Panx2 is an important regulator of the insulin secretory capacity and apoptosis in pancreatic ß-cells.


Subject(s)
Apoptosis/drug effects , Connexins/deficiency , Cytokines/pharmacology , Glucose Intolerance/metabolism , Insulin-Secreting Cells/metabolism , Animals , Connexins/metabolism , Gene Knockdown Techniques , Glucose Intolerance/pathology , Humans , Hyperglycemia/pathology , Inflammation/pathology , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects , Rats , STAT3 Transcription Factor/metabolism , Streptozocin
10.
Endocrinology ; 157(10): 3809-3820, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27479530

ABSTRACT

Increased expression of bone morphogenetic proteins (BMPs) in several tissues is associated with inflammation and type 2 diabetes mellitus. BMP2 and BMP4 mRNA expression is increased in pancreatic islets from db/db mice and ß-cell proliferation and function are inhibited by BMP4. The effect of BMPs on α-cells is currently unknown. Here, we investigate the effects of BMP4 on mouse and human α-cells in vitro. The effects of BMP4 on α-cell proliferation and function were investigated in islets isolated from male mice and from human donors, and in α-TC1-6 cells. The effects of BMP4 on α-cell function were assessed by determination of glucagon secretion and gene expression. Treatment with BMP4 for 24-96 hours inhibited glucagon secretion in a time-dependent manner in mouse and human islets. Glucagon content, preproglucagon and aristaless related homeobox mRNA expression were reduced after incubation with BMP4 in mouse islets, but not in human islets. The percentage of proliferating α-cells was reduced from 7.3 % to 0.2 % in mouse islets incubated with BMP4. α-cell proliferation in human islets ranged from 0 to 11.8 %, and BMP4 was found to inhibit proliferation of α-cells from all donors when proliferation was present. In agreement with the observations in primary islets, BMP4 decreased glucagon content, preproglucagon, and aristaless related homeobox mRNA expression in α-TC1-6 cells. Our findings suggest that BMP4 has an inhibitory role on glucagon secretion, α-cell growth, and expression of genes maintaining α-cell identity.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Glucagon-Secreting Cells/physiology , Adult , Animals , Apoptosis , Bone Morphogenetic Protein 2/metabolism , Cell Proliferation , Female , Gene Expression , Glucagon/metabolism , Humans , Male , Mice , Middle Aged , T-Lymphocytes, Cytotoxic/metabolism
11.
Diabetes ; 65(9): 2686-99, 2016 09.
Article in English | MEDLINE | ID: mdl-27284108

ABSTRACT

Pancreatic islet ß-cells are particularly susceptible to endoplasmic reticulum (ER) stress, which is implicated in ß-cell dysfunction and loss during the pathogenesis of type 1 diabetes (T1D). The peripheral membrane protein GAD65 is an autoantigen in human T1D. GAD65 synthesizes γ-aminobutyric acid, an important autocrine and paracrine signaling molecule and a survival factor in islets. We show that ER stress in primary ß-cells perturbs the palmitoylation cycle controlling GAD65 endomembrane distribution, resulting in aberrant accumulation of the palmitoylated form in trans-Golgi membranes. The palmitoylated form has heightened immunogenicity, exhibiting increased uptake by antigen-presenting cells and T-cell stimulation compared with the nonpalmitoylated form. Similar accumulation of GAD65 in Golgi membranes is observed in human ß-cells in pancreatic sections from GAD65 autoantibody-positive individuals who have not yet progressed to clinical onset of T1D and from patients with T1D with residual ß-cell mass and ongoing T-cell infiltration of islets. We propose that aberrant accumulation of immunogenic GAD65 in Golgi membranes facilitates inappropriate presentation to the immune system after release from stressed and/or damaged ß-cells, triggering autoimmunity.


Subject(s)
Autoantigens/metabolism , Autoimmunity/physiology , Endoplasmic Reticulum Stress/physiology , Glutamate Decarboxylase/metabolism , Golgi Apparatus/metabolism , Animals , Autoantibodies , Blotting, Western , Cell Line , Cells, Cultured , Diabetes Mellitus, Type 1/metabolism , Electrophoresis, Polyacrylamide Gel , Humans , Lipoylation , Pancreas/metabolism , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/metabolism
12.
J Diabetes Res ; 2016: 1312705, 2016.
Article in English | MEDLINE | ID: mdl-26962537

ABSTRACT

The relative contributions of the JNK subtypes in inflammatory ß-cell failure and apoptosis are unclear. The JNK protein family consists of JNK1, JNK2, and JNK3 subtypes, encompassing many different isoforms. INS-1 cells express JNK1α1, JNK1α2, JNK1ß1, JNK1ß2, JNK2α1, JNK2α2, JNK3α1, and JNK3α2 mRNA isoform transcripts translating into 46 and 54 kDa isoform JNK proteins. Utilizing Lentiviral mediated expression of shRNAs against JNK1, JNK2, or JNK3 in insulin-producing INS-1 cells, we investigated the role of individual JNK subtypes in IL-1ß-induced ß-cell apoptosis. JNK1 knockdown prevented IL-1ß-induced INS-1 cell apoptosis associated with decreased 46 kDa isoform JNK protein phosphorylation and attenuated Myc expression. Transient knockdown of Myc also prevented IL-1ß-induced apoptosis as well as caspase 3 cleavage. JNK2 shRNA potentiated IL-1ß-induced apoptosis and caspase 3 cleavage, whereas JNK3 shRNA did not affect IL-1ß-induced ß-cell death compared to nonsense shRNA expressing INS-1 cells. In conclusion, JNK1 mediates INS-1 cell death associated with increased Myc expression. These findings underline the importance of differentiated targeting of JNK subtypes in the development of inflammatory ß-cell failure and destruction.


Subject(s)
Apoptosis/drug effects , Insulin-Secreting Cells/drug effects , Insulin/metabolism , Interleukin-1beta/pharmacology , Mitogen-Activated Protein Kinase 8/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Caspase 3/metabolism , Cell Line, Tumor , Insulin Secretion , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Mice , Mitogen-Activated Protein Kinase 10/genetics , Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/metabolism , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , Rats , Signal Transduction/drug effects , Time Factors , Transfection
13.
Mol Cell Endocrinol ; 420: 24-36, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26610752

ABSTRACT

Interleukin-1ß (IL-1ß) and interferon-γ (IFNγ) contribute to type 1 diabetes (T1D) by inducing ß-cell death. Tumor necrosis factor (TNF) receptor-associated factor (TRAF) proteins are adaptors that transduce signaling from a variety of membrane receptors including cytokine receptors. We show here that IL-1ß and IFNγ upregulate the expression of TRAF2 in insulin-producing INS-1E cells and isolated rat pancreatic islets. siRNA-mediated knockdown (KD) of TRAF2 in INS-1E cells reduced IL-1ß-induced phosphorylation of JNK1/2, but not of p38 or ERK1/2 mitogen-activated protein kinases. TRAF2 KD did not modulate NFκB activation by cytokines, but reduced cytokine-induced inducible nitric oxide synthase (iNOS) promotor activity and expression. We further observed that IFNγ-stimulated phosphorylation of STAT3 required TRAF2. KD of TRAF2 or STAT3 reduced cytokine-induced caspase 3/7 activation, but, intriguingly, potentiated cytokine-mediated loss of plasma membrane integrity and augmented the number of propidium iodide-positive cells. Finally, we found that TRAF2 KD increased cytokine-induced production of reactive oxygen species (ROS). In summary, our data suggest that TRAF2 is an important mediator of IL-1ß and IFNγ signaling in pancreatic ß-cells.


Subject(s)
Insulin-Secreting Cells/cytology , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , STAT3 Transcription Factor/metabolism , TNF Receptor-Associated Factor 2/metabolism , Animals , Apoptosis/drug effects , Caspases/metabolism , Cell Line , Enzyme Activation/drug effects , Gene Knockdown Techniques , Humans , Inflammation Mediators/pharmacology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Mice , Necrosis , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects , Rats, Wistar , Reactive Oxygen Species/metabolism , Up-Regulation/drug effects
14.
J Interferon Cytokine Res ; 36(1): 20-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26308798

ABSTRACT

The proinflammatory cytokines interleukin-1 beta (IL-1ß) and interferon gamma (IFN-γ) play important roles in the progressive loss of beta-cell mass and function during development of both type 1 and type 2 diabetes. We have recently showed that bone morphogenetic protein (BMP)-2 and -4 are expressed in pancreatic islets and inhibit beta-cell growth and function. In this study, we describe that IL-1ß and IFN-γ induce the expression of BMP-2 suggesting a possible role for BMP-2 in mediating the effects of IL-1ß and IFN-γ on beta-cell apoptosis and dysfunction. IL-1ß increased BMP-2 mRNA levels 6- and 3-fold in isolated islets of Langerhans from neonatal rat and human. Downstream target genes of the BMP pathway were also increased by cytokine treatment and could be reversed by neutralization of endogenous BMP activity. Nuclear factor kappa B- (NFκB) binding sites were identified in the rat BMP-2 promoter, and reporter assays verified the role of NFκB in cytokine-induced BMP-2 expression. Electrophoretic mobility shift assay and chromatin immunoprecipitation assays confirmed NFκB binding to BMP-2 promoter upon IL-1ß stimulation in beta cells. In conclusion, we suggest that NFκB stimulates BMP-2 mRNA expression in rat and human beta cells upon cytokine exposure.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Insulin-Secreting Cells/drug effects , Interferon-gamma/metabolism , Interleukin-1beta/pharmacology , NF-kappa B/metabolism , RNA, Messenger/metabolism , Adult , Animals , Animals, Newborn , Binding Sites , Bone Morphogenetic Protein 2/genetics , Cell Line , Gene Expression Regulation , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Interferon-gamma/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Middle Aged , NF-kappa B/genetics , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , Rats , Rats, Wistar , Signal Transduction , Tissue Culture Techniques
15.
Peptides ; 69: 47-55, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25849341

ABSTRACT

AIMS/HYPOTHESIS: Combination treatment with exendin-4 and gastrin has proven beneficial in treatment of diabetes and preservation of beta cell mass in diabetic mice. Here, we examined the chronic effects of a GLP-1-gastrin dual agonist ZP3022 on glycemic control and beta cell dysfunction in overtly diabetic Zucker Diabetic Fatty (ZDF) rats. METHODS: ZDF rats aged 11 weeks were dosed s.c., b.i.d. for 8 weeks with vehicle, ZP3022, liraglutide, exendin-4, or gastrin-17 with or without exendin-4. Glycemic control was assessed by measurements of HbA1c and blood glucose levels, as well as glucose tolerance during an oral glucose tolerance test (OGTT). Beta cell dynamics were examined by morphometric analyses of beta and alpha cell fractions. RESULTS: ZP3022 improved glycemic control as measured by terminal HbA1c levels (6.2±0.12 (high dose) vs. 7.9±0.07% (vehicle), P<0.001), as did all treatments, except gastrin-17 monotherapy. In contrast, only ZP3022, exendin-4 and combination treatment with exendin-4 and gastrin-17 significantly improved glucose tolerance and increased insulin levels during an OGTT. Moreover, only ZP3022 significantly enhanced the beta cell fraction in ZDF rats, a difference of 41%, when compared to the vehicle group (0.31±0.03 vs. 0.22±0.02%, respectively, P<0.05). CONCLUSION: These data suggest that ZP3022 may have therapeutic potential in the prevention/delay of beta cell dysfunction in type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Gastrins/metabolism , Glucagon-Like Peptide 1/metabolism , Peptides/administration & dosage , Animals , Blood Glucose , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Gastrins/agonists , Glucagon-Like Peptide 1/agonists , Glycated Hemoglobin/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Mice , Rats , Rats, Zucker
16.
Diabetologia ; 58(6): 1282-90, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25828920

ABSTRACT

AIMS/HYPOTHESIS: Type 2 diabetes is characterised by progressive loss of pancreatic beta cell mass and function. Therefore, it is of therapeutic interest to identify factors with the potential to improve beta cell proliferation and insulin secretion. Bone morphogenetic protein 4 (BMP4) expression is increased in diabetic animals and BMP4 reduces glucose-stimulated insulin secretion (GSIS). Here, we investigate the molecular mechanism behind this inhibition. METHODS: BMP4-mediated inhibition of GSIS was investigated in detail using single cell electrophysiological measurements and live cell Ca(2+) imaging. BMP4-mediated gene expression changes were investigated by microarray profiling, quantitative PCR and western blotting. RESULTS: Prolonged exposure to BMP4 reduced GSIS from rodent pancreatic islets. This inhibition was associated with decreased exocytosis due to a reduced Ca(2+) current through voltage-dependent Ca(2+) channels. To identify proteins involved in the inhibition of GSIS, we investigated global gene expression changes induced by BMP4 in neonatal rat pancreatic islets. Expression of the Ca(2+)-binding protein calbindin1 was significantly induced by BMP4. Overexpression of calbindin1 in primary islet cells reduced GSIS, and the effect of BMP4 on GSIS was lost in islets from calbindin1 (Calb1) knockout mice. CONCLUSIONS/INTERPRETATION: We found BMP4 treatment to markedly inhibit GSIS from rodent pancreatic islets in a calbindin1-dependent manner. Calbindin1 is suggested to mediate the effect of BMP4 by buffering Ca(2+) and decreasing Ca(2+) channel activity, resulting in diminished insulin exocytosis. Both BMP4 and calbindin1 are potential pharmacological targets for the treatment of beta cell dysfunction.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Calbindin 1/metabolism , Calcium/metabolism , Insulin-Secreting Cells/cytology , Insulin/metabolism , Animals , Calbindin 1/genetics , Electrophysiological Phenomena , Female , Gene Expression Profiling , Gene Expression Regulation , Insulin Secretion , Islets of Langerhans/cytology , Male , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Rats , Rats, Wistar , Up-Regulation
17.
PLoS One ; 10(2): e0117130, 2015.
Article in English | MEDLINE | ID: mdl-25647668

ABSTRACT

The inhibitory neurotransmitter GABA is synthesized by the enzyme glutamic acid decarboxylase (GAD) in neurons and in pancreatic ß-cells in islets of Langerhans where it functions as a paracrine and autocrine signaling molecule regulating the function of islet endocrine cells. The localization of the two non-allelic isoforms GAD65 and GAD67 to vesicular membranes is important for rapid delivery and accumulation of GABA for regulated secretion. While the membrane anchoring and trafficking of GAD65 are mediated by intrinsic hydrophobic modifications, GAD67 remains hydrophilic, and yet is targeted to vesicular membrane pathways and synaptic clusters in neurons by both a GAD65-dependent and a distinct GAD65-independent mechanism. Herein we have investigated the membrane association and targeting of GAD67 and GAD65 in monolayer cultures of primary rat, human, and mouse islets and in insulinoma cells. GAD65 is primarily detected in Golgi membranes and in peripheral vesicles distinct from insulin vesicles in ß-cells. In the absence of GAD65, GAD67 is in contrast primarily cytosolic in ß-cells; its co-expression with GAD65 is necessary for targeting to Golgi membranes and vesicular compartments. Thus, the GAD65-independent mechanism for targeting of GAD67 to synaptic vesicles in neurons is not functional in islet ß-cells. Therefore, only GAD65:GAD65 homodimers and GAD67:GAD65 heterodimers, but not the GAD67:GAD67 homodimer gain access to vesicular compartments in ß-cells to facilitate rapid accumulation of newly synthesized GABA for regulated secretion and fine tuning of GABA-signaling in islets of Langerhans.


Subject(s)
Glutamate Decarboxylase/metabolism , Insulin-Secreting Cells/metabolism , Neurons/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Cells, Cultured , Glutamate Decarboxylase/analysis , Golgi Apparatus/metabolism , Humans , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Synaptic Vesicles/metabolism
18.
J Appl Toxicol ; 35(7): 842-50, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25413577

ABSTRACT

There is a medical need for new insulin analogues. Yet, molecular alterations to the insulin molecule can theoretically result in analogues with carcinogenic effects. Preclinical carcinogenicity risk assessment for insulin analogues rests to a large extent on mitogenicity assays in cell lines. We therefore optimized mitogenicity assay conditions for a panel of five cell lines. All cell lines expressed insulin receptors (IR), IGF-I receptors (IGF-IR) and hybrid receptors, and in all cell lines, insulin as well as the comparator compounds X10 and IGF-I caused phosphorylation of the IR as well as IGF-IR. Insulin exhibited mitogenicity EC(50) values in the single-digit nanomolar to picomolar range. We observed correlations across cell types between (i) mitogenic potency of insulin and IGF-IR/IR ratio, (ii) Akt phosphorylation and mitogenic potency and (iii) Akt phosphorylation and IR phosphorylation. Using siRNA-mediated knockdown of IR and IGF-IR, we observed that in HCT 116 cells the IR appeared dominant in driving the mitogenic response to insulin, whereas in MCF7 cells the IGF-IR appeared dominant in driving the mitogenic response to insulin. Together, our results show that the IR as well as IGF-IR may contribute to the mitogenic potency of insulin. While insulin was a more potent mitogen than IGF-I in cells expressing more IR than IGF-IR, the hyper-mitogenic insulin analogue X10 was a more potent mitogen than insulin across all cell types, supporting that the hyper-mitogenic effect of X10 involves the IR as well as the IGF-IR. These results are relevant for preclinical safety assessment of developmental insulin analogues.


Subject(s)
Insulin/pharmacology , Mitosis/drug effects , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme-Linked Immunosorbent Assay , HCT116 Cells/drug effects , Hep G2 Cells/drug effects , Humans , Insulin/analogs & derivatives , MCF-7 Cells/drug effects , Rats
19.
J Mol Endocrinol ; 53(3): 417-27, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25324488

ABSTRACT

During the development of diabetes ß-cells are exposed to elevated concentrations of proinflammatory cytokines, TNFα and IL1ß, which in vitro induce ß-cell death. The class B G-protein-coupled receptors (GPCRs): corticotropin-releasing factor receptor 1 (CRFR1) and CRFR2 are expressed in pancreatic islets. As downstream signaling by other class B GPCRs can protect against cytokine-induced ß-cell apoptosis, we evaluated the protective potential of CRFR activation in ß-cells in a pro-inflammatory setting. CRFR1/CRFR2 ligands activated AKT and CRFR1 signaling and reduced apoptosis in human islets. In rat and mouse insulin-secreting cell lines (INS-1 and MIN6), CRFR1 agonists upregulated insulin receptor substrate 2 (IRS2) expression, increased AKT activation, counteracted the cytokine-mediated decrease in BAD phosphorylation, and inhibited apoptosis. The anti-apoptotic signaling was dependent on prolonged exposure to corticotropin-releasing factor family peptides and followed PKA-mediated IRS2 upregulation. This indicates that CRFR signaling counteracts proinflammatory cytokine-mediated apoptotic pathways through upregulation of survival signaling in ß-cells. Interestingly, CRFR signaling also counteracted basal apoptosis in both cultured INS-1 cells and intact human islets.


Subject(s)
Apoptosis/drug effects , Corticotropin-Releasing Hormone/pharmacology , Cytokines/adverse effects , Cytoprotection/drug effects , Insulin-Secreting Cells/drug effects , Receptors, Corticotropin-Releasing Hormone/agonists , Animals , Cell Death/drug effects , Cells, Cultured , Humans , Insulin-Secreting Cells/physiology , Interleukin-1beta/adverse effects , Mice , Rats , Tumor Necrosis Factor-alpha/adverse effects
20.
Diabetologia ; 57(12): 2546-54, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25260823

ABSTRACT

AIMS/HYPOTHESIS: Impairment of beta cell mass and function is evident in both type 1 and type 2 diabetes. In healthy physiological conditions pancreatic beta cells adapt to the body's increasing insulin requirements by proliferation and improved function. We hypothesised that during the development of diabetes, there is an increase in the expression of inhibitory factors that prevent the beta cells from adapting to the increased need for insulin. We evaluated the effects of bone morphogenetic protein (BMP) 2 and -4 on beta cells. METHODS: The effects of BMP2 and -4 on beta cell proliferation, apoptosis, gene expression and insulin release were studied in isolated islets of Langerhans from rats, mice and humans. The expression of BMPs was analysed by immunocytochemistry and real-time PCR. The role of endogenous BMP was investigated using a soluble and neutralising form of the BMP receptor 1A. RESULTS: BMP2 and -4 were found to inhibit basal as well as growth factor-stimulated proliferation of primary beta cells from rats and mice. Bmp2 and Bmp4 mRNA and protein were expressed in islets and regulated by inflammatory cytokines. Neutralisation of endogenous BMP activity resulted in enhanced proliferation of rodent beta cells. The expression of Id mRNAs was induced by BMP4 in rat and human islets. Finally, glucose-induced insulin secretion was significantly impaired in rodent and human islets pre-treated with BMP4, and inhibition of BMP activity resulted in enhanced insulin release. CONCLUSIONS/INTERPRETATION: These data show that BMP2 and -4 exert inhibitory actions on beta cells in vitro and suggest that BMPs exert regulatory roles of beta cell growth and function.


Subject(s)
Apoptosis/drug effects , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 4/pharmacology , Cell Proliferation/drug effects , Gene Expression/drug effects , Insulin-Secreting Cells/drug effects , Animals , Cells, Cultured , Insulin/metabolism , Insulin-Secreting Cells/physiology , Mice , Rats , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...