Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Hum Mol Genet ; 24(10): 3004, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25753258

ABSTRACT

In this article, Figure 2F was incorrect. The correct panel is shown below. The authors sincerely apologise for this error.

2.
Biochem J ; 451(2): 227-34, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23390957

ABSTRACT

The incretins, GIP (glucose-dependent insulinotropic polypeptide) and GLP-1 (glucagon-like peptide-1) are gastrointestinal hormones conferring a number of beneficial effects on ß-cell secretion, survival and proliferation. In a previous study, it was demonstrated that delayed rectifier channel protein Kv2.1 contributes to ß-cell apoptosis and that the prosurvival effects of incretins involve Kv2.1 PTMs (post-translational modifications), including phosphorylation and acetylation. Since Kv1.5 overexpression was also shown to stimulate ß-cell death, the present study was initiated in order to determine whether incretins modulate Kv1.5α-Kvß2 interaction via PTM and the mechanisms involved. GIP and GLP-1 reduced apoptosis in INS-1 ß-cells (clone 832/13) overexpressing Kv1.5, and RNAi (RNA interference)-mediated knockdown of endogenous Kv1.5 attenuated apoptotic ß-cell death. Both GIP and GLP-1 increased phosphorylation and acetylation of Kv1.5 and its Kvß2 protein subunit, leading to their enhanced interaction. Further studies demonstrated that CBP [CREB (cAMP-response-element-binding protein)-binding protein]/SirT1 mediated acetylation/deacetylation and interaction between Kvß2 and Kv1.5 in response to GIP or GLP-1. Incretin regulation of ß-cell function therefore involves the acetylation of multiple Kvα and Kvß subunits.


Subject(s)
CREB-Binding Protein/metabolism , Incretins/pharmacology , Insulin-Secreting Cells/metabolism , Kv1.5 Potassium Channel/metabolism , Potassium Channels, Voltage-Gated/metabolism , Sirtuin 1/metabolism , Acetylation/drug effects , Apoptosis/drug effects , CREB-Binding Protein/genetics , Cell Survival/drug effects , Cells, Cultured , Gastric Inhibitory Polypeptide/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Gene Knockdown Techniques , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/pharmacology , Humans , Incretins/metabolism , Insulin-Secreting Cells/drug effects , Kv1.5 Potassium Channel/genetics , Phosphorylation/drug effects , Protein Processing, Post-Translational , Shaker Superfamily of Potassium Channels
3.
Diabetes ; 62(2): 471-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23002036

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) is an incretin hormone that also plays a regulatory role in fat metabolism. In 3T3-L1 cells, resistin was demonstrated to be a key mediator of GIP stimulation of lipoprotein lipase (LPL) activity, involving activation of protein kinase B (PKB) and reduced phosphorylation of liver kinase B1 (LKB1) and AMP-activated protein kinase (AMPK). The current study was initiated to determine whether resistin has additional roles in GIP-regulated adipocyte functions. Analysis of primary adipocytes isolated from Retn(-/-), Retn(+/-), and Retn(+/+) mice found that GIP stimulated the PKB/LKB1/AMPK/LPL pathway and fatty acid uptake only in Retn(+/+) adipocytes, suggesting that GIP signaling and/or GIP responsiveness were compromised in Retn(+/-) and Retn(-/-) adipocytes. GIP receptor (GIPR) protein and mRNA were decreased in Retn(+/-) and Retn(-/-) adipocytes, but resistin treatment rescued LPL responsiveness to GIP. In addition, genes encoding tumor necrosis factor (TNF), TNF receptor 2 (TNFR2), and the signaling proteins stress-activated protein kinase (SAPK)/Jun NH(2)-terminal kinase (JNK), were downregulated, and phosphorylated levels of SAPK/JNK/c-Jun were decreased in Retn(-/-) mice. Chromatin immunoprecipitation assays were used to identify a 12-O-tetradecanoylphorbol-13-acetate (TPA)-response element (TRE-III) responsible for c-Jun-mediated transcriptional activation of Gipr. Blunted GIP responsiveness in Retn(+/-) and Retn(-/-) adipocytes was therefore largely due to the greatly reduced GIPR expression associated with decreased c-Jun-mediated transcriptional activation of Gipr.


Subject(s)
Adipocytes/metabolism , Gastric Inhibitory Polypeptide/metabolism , Receptors, Gastrointestinal Hormone/biosynthesis , Resistin/metabolism , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipocytes/drug effects , Animals , Carcinogens/pharmacology , Cells, Cultured , Fatty Acids/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Gene Expression Regulation , Lipoprotein Lipase/metabolism , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 8/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/pharmacology , Receptors, Tumor Necrosis Factor, Type II/biosynthesis , Receptors, Tumor Necrosis Factor, Type II/genetics , Resistin/genetics , Resistin/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology
4.
PLoS One ; 7(7): e40156, 2012.
Article in English | MEDLINE | ID: mdl-22802954

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) is a gastrointestinal hormone that potentiates glucose-stimulated insulin secretion during a meal. Since GIP has also been shown to exert ß-cell prosurvival and adipocyte lipogenic effects in rodents, both GIP receptor agonists and antagonists have been considered as potential therapeutics in type 2 diabetes (T2DM). In the present study, we tested the hypothesis that chronically elevating GIP levels in a transgenic (Tg) mouse model would increase adipose tissue expansion and exert beneficial effects on glucose homeostasis. In contrast, although GIP Tg mice demonstrated enhanced ß-cell function, resulting in improved glucose tolerance and insulin sensitivity, they exhibited reduced diet-induced obesity. Adipose tissue macrophage infiltration and hepatic steatosis were both greatly reduced, and a number of genes involved in lipid metabolism/inflammatory signaling pathways were found to be down-regulated. Reduced adiposity in GIP Tg mice was associated with decreased energy intake, involving overexpression of hypothalamic GIP. Together, these studies suggest that, in the context of over-nutrition, transgenic GIP overexpression has the potential to improve hepatic and adipocyte function as well as glucose homeostasis.


Subject(s)
Fatty Liver/prevention & control , Gastric Inhibitory Polypeptide/biosynthesis , Glucose/metabolism , Homeostasis , Obesity/etiology , Adipose Tissue/metabolism , Animals , Diet, High-Fat , Energy Metabolism , Gastric Inhibitory Polypeptide/pharmacology , Glucose Intolerance/metabolism , Glucose Tolerance Test , Insulin Resistance/physiology , Male , Mice , Mice, Transgenic , Obesity/metabolism
5.
PLoS One ; 7(3): e32675, 2012.
Article in English | MEDLINE | ID: mdl-22412906

ABSTRACT

In response to a meal, Glucose-dependent Insulinotropic Polypeptide (GIP) and Glucagon-like Peptide-1 (GLP-1) are released from gut endocrine cells into the circulation and interact with their cognate G-protein coupled receptors (GPCRs). Receptor activation results in tissue-selective pleiotropic responses that include augmentation of glucose-induced insulin secretion from pancreatic beta cells. N-glycosylation and receptor oligomerization are co-translational processes that are thought to regulate the exit of functional GPCRs from the ER and their maintenance at the plasma membrane. Despite the importance of these regulatory processes, their impact on functional expression of GIP and GLP-1 receptors has not been well studied. Like many family B GPCRs, both the GIP and GLP-1 receptors possess a large extracellular N-terminus with multiple consensus sites for Asn-linked (N)-glycosylation. Here, we show that each of these Asn residues is glycosylated when either human receptor is expressed in Chinese hamster ovary cells. N-glycosylation enhances cell surface expression and function in parallel but exerts stronger control over the GIP receptor than the GLP-1 receptor. N-glycosylation mainly lengthens receptor half-life by reducing degradation in the endoplasmic reticulum. N-glycosylation is also required for expression of the GIP receptor at the plasma membrane and efficient GIP potentiation of glucose-induced insulin secretion from the INS-1 pancreatic beta cell line. Functional expression of a GIP receptor mutant lacking N-glycosylation is rescued by co-expressed wild type GLP1 receptor, which, together with data obtained using Bioluminescence Resonance Energy Transfer, suggests formation of a GIP-GLP1 receptor heteromer.


Subject(s)
Protein Multimerization , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/metabolism , Amino Acid Sequence , Animals , Cell Line , Chlorocebus aethiops , Consensus Sequence , Cyclic AMP/biosynthesis , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Glucagon-Like Peptide-1 Receptor , Glycosylation , Humans , Insulin/metabolism , Insulin Secretion , Ligands , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Molecular Sequence Data , Mutation , Protein Isoforms , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Gastrointestinal Hormone/genetics , Receptors, Glucagon/chemistry , Receptors, Glucagon/genetics , Sequence Alignment
6.
J Lipid Res ; 52(4): 759-70, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21245029

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) is a gastrointestinal hormone that exerts insulinotropic and growth and survival effects on pancreatic ß-cells. Additionally, there is increasing evidence supporting an important role for GIP in the regulation of adipocyte metabolism. In the current study we examined the molecular mechanisms involved in the regulation of GIP receptor (GIPR) expression in 3T3-L1 cells. GIP acted synergistically with insulin to increase neutral lipid accumulation during progression of 3T3-L1 preadipocytes to the adipocyte phenotype. Both GIPR protein and mRNA expression increased during 3T3-L1 cell differentiation, and this increase was associated with upregulation of nuclear levels of sterol response element binding protein 1c (SREBP-1c) and peroxisome proliferator-activated receptor γ (PPARγ), as well as acetylation of histones H3/H4. The PPARγ receptor agonists LY171883 and rosiglitazone increased GIPR expression in differentiated 3T3-L1 adipocytes, whereas the antagonist GW9662 ablated expression. Additionally, both PPARγ and acetylated histones H3/H4 were shown to bind to a region of the GIPR promoter containing the peroxisome proliferator response element (PPRE). Knockdown of PPARγ in differentiated 3T3-L1 adipocytes, using RNA interference, reduced GIPR expression, supporting a functional regulatory role. Taken together, these studies show that GIP and insulin act in a synergistic manner on 3T3-L1 cell development and that adipocyte GIPR expression is upregulated through a mechanism involving interactions between PPARγ and a GIPR promoter region containing an acetylated histone region.


Subject(s)
Adipocytes/metabolism , Histones/metabolism , PPAR gamma/metabolism , Receptors, Gastrointestinal Hormone/metabolism , 3T3-L1 Cells , Acetylation , Adipocytes/cytology , Animals , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Chromatin Immunoprecipitation , Immunoprecipitation , Mice , PPAR gamma/genetics , Promoter Regions, Genetic , Protein Binding , RNA Interference , Receptors, Gastrointestinal Hormone/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism
7.
Vitam Horm ; 84: 21-79, 2010.
Article in English | MEDLINE | ID: mdl-21094896

ABSTRACT

The insulin secretory response to a meal results largely from glucose stimulation of the pancreatic islets and both direct and indirect (autonomic) glucose-dependent stimulation by incretin hormones released from the gastrointestinal tract. Two incretins, Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), have so far been identified. Localization of the cognate G protein-coupled receptors for GIP and GLP-1 revealed that they are present in numerous tissues in addition to the endocrine pancreas, including the gastrointestinal, cardiovascular, central nervous and autonomic nervous systems (ANSs), adipose tissue, and bone. At these sites, the incretin hormones exert a range of pleiotropic effects, many of which contribute to the integration of processes involved in the regulation of food intake, and nutrient and mineral processing and storage. From detailed studies at the cellular and molecular level, it is also evident that both incretin hormones act via multiple signal transduction pathways that regulate both acute and long-term cell function. Here, we provide an overview of current knowledge relating to the physiological roles of GIP and GLP-1, with specific emphasis on their modes of action on islet hormone secretion, ß-cell proliferation and survival, central and autonomic neuronal function, gastrointestinal motility, and glucose and lipid metabolism. However, it is emphasized that despite intensive research on the various body systems, in many cases there is uncertainty as to the pathways by which the incretins mediate their pleiotropic effects and only a rudimentary understanding of the underlying cellular mechanisms involved, and these are challenges for the future.


Subject(s)
Gastric Inhibitory Polypeptide/physiology , Glucagon-Like Peptide 1/physiology , Insulin/physiology , Islets of Langerhans/physiology , Animals , Humans , Insulin/metabolism , Insulin Secretion , Receptors, Glucagon/physiology
8.
J Lipid Res ; 51(11): 3145-57, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20693566

ABSTRACT

GIP (glucose-dependent insulinotropic polypeptide) is a gastrointestinal hormone that regulates pancreatic islet function. Additionally, emerging evidence suggests an important physiological role for GIP in the regulation of adipocyte metabolism. In previous studies on the lipogenic effects of GIP, it was shown to increase adipocyte lipoprotein lipase (LPL) activity in both differentiated 3T3-L1 cells and human adipocytes through a pathway involving activation of protein kinase B (PKB)/Akt. In the current study, we examined the effects of GIP on LPL gene expression. GIP in the presence of insulin increased LPL gene expression in human adipocytes and LPL promoter activity in GIP receptor-expressing HEK-293 cells, and both effects were greatly reduced by the transcription inhibitor actinomycin D. Subsequent studies established that GIP increased phosphorylation of Serine 133 in cAMP-response element binding protein (CREB) and the nuclear localization of cAMP-responsive CREB coactivator 2 (TORC2) through a pathway involving phosphatidylinositol 3-kinase (PI3-K), PKB, and AMP-activated protein kinase (AMPK). However, in the presence of insulin, GIP failed to activate the cAMP/PKA pathway. Knockdown of CREB and TORC2 using RNA interference reduced LPL expression, supporting a functional regulatory role. GIP-induced phospho-CREB and TORC2 were shown to bind to a cAMP-response element (-II) site in the human LPL promoter and GIP increased protein-protein interactions of these two factors. The lipogenic effects of GIP in the presence of insulin are therefore at least partially mediated by upregulation of adipocyte LPL gene transcription through a pathway involving PI3-K/PKB/AMPK-dependent CREB/TORC2 activation.


Subject(s)
Adipocytes/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Lipoprotein Lipase/genetics , Transcription Factors/metabolism , Transcriptional Activation/drug effects , AMP-Activated Protein Kinases/metabolism , Active Transport, Cell Nucleus/drug effects , Adipocytes/cytology , Adipocytes/enzymology , Adipocytes/metabolism , Adult , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Female , HEK293 Cells , Humans , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-akt/metabolism , Transcription, Genetic/drug effects
9.
Diabetes ; 59(7): 1739-50, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20368408

ABSTRACT

OBJECTIVE: Treatment of NOD mice with the dipeptidyl peptidase-IV (DPP-IV) inhibitor sitagliptin preserved islet transplants through a pathway involving modulation of splenic CD4(+) T-cell migration. In the current study, effects of sitagliptin on migration of additional subsets of CD4(+) T-cells were examined and underlying molecular mechanisms were further defined. RESEARCH DESIGN AND METHODS: Effects of sitagliptin on migration of NOD mouse splenic, thymic, and lymph node CD4(+) T-cells were determined. Signaling modules involved in DPP-IV-, Sitagliptin- and incretin-mediated modulation of CD4(+) T-cell migration were studied using Western blot and Rac1 and nuclear factor-kappaB (NF-kappaB) activity assays. RESULTS: Migration of splenic and lymph node CD4(+) T-cells of diabetic NOD mice was reduced by sitagliptin treatment. In vitro treatment of splenic, but not thymic or lymph node CD4(+) T-cells, from nondiabetic NOD mice with soluble (s) DPP-IV increased migration. Sitagliptin abolished sDPP-IV effects on splenic CD4(+) T-cell migration, whereas incretins decreased migration of lymph node, but not splenic, CD4(+) T-cells. Splenic CD4(+) T-cells demonstrating increased in vitro migration in response to sDPP-IV and lymph node CD4(+) T-cells that were nonresponsive to incretins selectively infiltrated islets of NOD mice, after injection. Sitagliptin decreases migration of splenic CD4(+) T-cells through a pathway involving Rac1/vasodilator-stimulated phosphoprotein, whereas its inhibitory effects on the migration of lymph node CD4(+) T-cells involve incretin-activation of the NF-kappaB pathway. CONCLUSIONS: Benefits of sitagliptin treatment in diabetic NOD mice may be mediated through selective effects on subpopulations of T-cells that are related to autoimmunity.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Cell Movement/drug effects , Dipeptidyl Peptidase 4/metabolism , Incretins/metabolism , Pyrazines/pharmacology , T-Lymphocyte Subsets/drug effects , Triazoles/pharmacology , Analysis of Variance , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Movement/immunology , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Mice, Inbred NOD , Signal Transduction/drug effects , Signal Transduction/immunology , Sitagliptin Phosphate , Spleen/drug effects , Spleen/immunology , Spleen/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/drug effects , Thymus Gland/immunology , Thymus Gland/metabolism
10.
PLoS One ; 5(3): e9590, 2010 Mar 09.
Article in English | MEDLINE | ID: mdl-20231880

ABSTRACT

AIMS: The gastrointestinal hormone GIP promotes pancreatic islet function and exerts pro-survival actions on cultured beta-cells. However, GIP also promotes lipogenesis, thus potentially restricting its therapeutic use. The current studies evaluated the effects of a truncated GIP analog, D-Ala(2)-GIP(1-30) (D-GIP(1-30)), on glucose homeostasis and beta-cell mass in rat models of diabetes. MATERIALS AND METHODS: The insulinotropic and pro-survival potency of D-GIP(1-30) was evaluated in perfused pancreas preparations and cultured INS-1 beta-cells, respectively, and receptor selectivity evaluated using wild type and GIP receptor knockout mice. Effects of D-GIP(1-30) on beta-cell function and glucose homeostasis, in vivo, were determined using Lean Zucker rats, obese Vancouver diabetic fatty rats, streptozotocin treated rats, and obese Zucker diabetic fatty rats, with effects on beta-cell mass determined in histological studies of pancreatic tissue. Lipogenic effects of D-GIP(1-30) were evaluated on cultured 3T3-L1 adipocytes. RESULTS: Acutely, D-GIP(1-30) improved glucose tolerance and insulin secretion. Chronic treatment with D-GIP(1-30) reduced levels of islet pro-apoptotic proteins in Vancouver diabetic fatty rats and preserved beta-cell mass in streptozotocin treated rats and Zucker diabetic fatty rats, resulting in improved insulin responses and glycemic control in each animal model, with no change in body weight. In in vitro studies, D-GIP(1-30) exhibited equivalent potency to GIP(1-42) on beta-cell function and survival, but greatly reduced action on lipoprotein lipase activity in 3T3-L1 adipocytes. CONCLUSIONS: These findings demonstrate that truncated forms of GIP exhibit potent anti-diabetic actions, without pro-obesity effects, and that the C-terminus contributes to the lipogenic actions of GIP.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Apoptosis , Diabetes Mellitus, Experimental/drug therapy , Insulin-Secreting Cells/metabolism , Receptors, Gastrointestinal Hormone/agonists , 3T3 Cells , Adipocytes/cytology , Animals , Glucose/metabolism , Glucose Tolerance Test , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Zucker , Receptors, Gastrointestinal Hormone/chemistry
11.
J Biol Chem ; 284(44): 30372-82, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19748889

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) potentiates glucose-stimulated insulin secretion, insulin biosynthesis, and beta-cell proliferation and survival. In previous studies GIP was shown to promote beta-cell survival by modulating the activity of multiple signaling modules and regulating gene transcription of pro- and anti-apoptotic bcl-2 family proteins. We have now evaluated the mechanisms by which GIP regulates the dynamic interactions between cytoplasmic bcl-2 family members and the mitochondria in INS-1 cells during apoptosis induced by treatment with staurosporine (STS), an activator of the mitochondria-mediated apoptotic pathway. STS induced translocation of bad and bimEL, activation of mitochondrial bax, release of mitochondrial cytochrome c, cleavage of caspase-3, and apoptosis. Each response was significantly diminished by GIP. Using selective enzyme inhibitors, overexpression of dominant-negative Akt, and Akt siRNA, it was demonstrated that GIP promoted beta-cell survival via Akt-dependent suppression of p38 MAPK and JNK and that combined inhibition was sufficient to explain the entire pro-survival responses to GIP during STS treatment. This signaling pathway also explained the pro-survival effects of GIP on INS-1 cells exposed to two other promoters of stress: thapsigargin (endoplasmic reticulum stress) and etoposide (genotoxic stress). Importantly, we discovered that GIP suppressed p38 MAPK and JNK via Akt-mediated changes in the phosphorylation state of the apoptosis signal-regulating kinase 1 in INS-1 cells and human islets, resulting in inhibition of its activity. Inhibition of apoptosis by GIP is therefore mediated via a key pathway involving Akt-dependent inhibition of apoptosis signal-regulating kinase 1, which subsequently prevents the pro-apoptotic actions of p38 MAPK and JNK.


Subject(s)
Gastric Inhibitory Polypeptide/physiology , Insulin-Secreting Cells/cytology , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/physiology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Apoptosis , Cell Survival , Cells, Cultured , Humans , Islets of Langerhans/cytology , Signal Transduction/physiology
12.
Am J Physiol Endocrinol Metab ; 297(3): E695-707, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19602585

ABSTRACT

In addition to its primary role in regulating glucose production from the liver, glucagon has many other actions, reflected by the wide tissue distribution of the glucagon receptor (Gcgr). To investigate the role of glucagon in the regulation of insulin secretion and whole body glucose homeostasis in vivo, we generated mice overexpressing the Gcgr specifically on pancreatic beta-cells (RIP-Gcgr). In vivo and in vitro insulin secretion in response to glucagon and glucose was increased 1.7- to 3.9-fold in RIP-Gcgr mice compared with controls. Consistent with the observed increase in insulin release in response to glucagon and glucose, the glucose excursion resulting from both a glucagon challenge and intraperitoneal glucose tolerance test (IPGTT) was significantly reduced in RIP-Gcgr mice compared with controls. However, RIP-Gcgr mice display similar glucose responses to an insulin challenge. beta-Cell mass and pancreatic insulin content were also increased (20 and 50%, respectively) in RIP-Gcgr mice compared with controls. When fed a high-fat diet (HFD), both control and RIP-Gcgr mice developed similar degrees of obesity and insulin resistance. However, the severity of both fasting hyperglycemia and impaired glucose tolerance (IGT) were reduced in RIP-Gcgr mice compared with controls. Furthermore, the insulin response of RIP-Gcgr mice to an IPGTT was twice that of controls when fed the HFD. These data indicate that increased pancreatic beta-cell expression of the Gcgr increased insulin secretion, pancreatic insulin content, beta-cell mass, and, when mice were fed a HFD, partially protected against hyperglycemia and IGT.


Subject(s)
Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Receptors, Glucagon/genetics , Animals , Cell Proliferation , Cell Size , Cells, Cultured , Diet, Atherogenic , Female , Glucose Intolerance/genetics , Hyperglycemia/genetics , Insulin/metabolism , Insulin Secretion , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Organ Specificity/genetics , Receptors, Glucagon/metabolism , Transfection
13.
Hum Mol Genet ; 18(13): 2388-99, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19386626

ABSTRACT

Recent human genetics studies have revealed that common variants of the TCF7L2 (T-cell factor 7-like 2, formerly known as TCF4) gene are strongly associated with type 2 diabetes mellitus (T2DM). We have shown that TCF7L2 expression in the beta-cells is correlated with function and survival of the insulin-producing pancreatic beta-cell. In order to understand how variations in TCF7L2 influence diabetes progression, we investigated its mechanism of action in the beta-cell. We show robust differences in TCF7L2 expression between healthy controls and models of T2DM. While mRNA levels were approximately 2-fold increased in isolated islets from the diabetic db/db mouse, the Vancouver Diabetic Fatty (VDF) Zucker rat and the high fat/high sucrose diet-treated mouse compared with the non-diabetic controls, protein levels were decreased. A similar decrease was observed in pancreatic sections from patients with T2DM. In parallel, expression of the receptors for glucagon-like peptide 1 (GLP-1R) and glucose-dependent insulinotropic polypeptide (GIP-R) was decreased in islets from humans with T2DM as well as in isolated human islets treated with siRNA to TCF7L2 (siTCF7L2). Also, insulin secretion stimulated by glucose, GLP-1 and GIP, but not KCl or cyclic adenosine monophosphate (cAMP) was impaired in siTCF7L2-treated isolated human islets. Loss of TCF7L2 resulted in decreased GLP-1 and GIP-stimulated AKT phosphorylation, and AKT-mediated Foxo-1 phosphorylation and nuclear exclusion. Our findings suggest that beta-cell function and survival are regulated through an interplay between TCF7L2 and GLP-1R/GIP-R expression and signaling in T2DM.


Subject(s)
Down-Regulation , Insulin-Secreting Cells/physiology , Receptors, Gastrointestinal Hormone/genetics , Receptors, Glucagon/genetics , TCF Transcription Factors/metabolism , Aged , Animals , Female , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor , Humans , In Vitro Techniques , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/physiology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Rats , Rats, Zucker , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/metabolism , Signal Transduction , TCF Transcription Factors/genetics , Transcription Factor 7-Like 2 Protein
14.
J Biol Chem ; 284(19): 12896-904, 2009 May 08.
Article in English | MEDLINE | ID: mdl-19279000

ABSTRACT

Chromatin can exert a regulatory effect on gene transcription by modulating the access of transcription factors to target genes. In the present study, we examined whether nuclear actions of the incretin hormones, glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1, involve modulation of beta-cell chromatin structure. Stimulation of INS-1(832/13) beta-cells or dispersed mouse islets with glucose-dependent insulinotropic polypeptide or glucagon-like peptide-1 resulted in the post-translational modification of core H3 histones, through acetylation and phosphorylation. Both increased histone H3 acetyltransferase and reduced histone deacetylase activities contributed. Subsequent studies demonstrated that incretin-mediated histone H3 modifications involved activation of protein kinase A, p42/44 mitogen-activated protein kinase (MAPK), and p38 MAPK signaling modules, resulting in the activation of mitogen- and stress-activated kinase-1. Additionally, modification of histone H3 increased its association with the transcription factor, phosphorylated cAMP-response element-binding protein (phospho-CREB) and with cAMP-responsive CREB coactivator 2. Incretin-activated CREB-related Bcl-2 transcription was greatly reduced by a histone acetyltransferase inhibitor, demonstrating the functional importance of histone H3 modification. This appears to be the first demonstration of beta-cell chromatin modification in response to the incretins and the studies indicate that their regulatory effects involve coordinated nuclear interactions between specific signaling modules, chromatin-modifying enzymes and transcription factors.


Subject(s)
Chromatin/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Glucagon-Like Peptide 1/pharmacology , Glucose/metabolism , Insulin-Secreting Cells/drug effects , Acetylation , Animals , Blotting, Western , Cell Nucleus/metabolism , Chromatin/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Histone Deacetylase 1 , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/genetics , Histones/metabolism , Immunoprecipitation , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Processing, Post-Translational , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction , Trans-Activators/metabolism , Transcription Factors
15.
Vitam Horm ; 80: 409-71, 2009.
Article in English | MEDLINE | ID: mdl-19251046

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP; gastric inhibitory polypeptide) is a 42 amino acid hormone that is produced by enteroendocrine K-cells and released into the circulation in response to nutrient stimulation. Both GIP and glucagon-like peptide-1 (GLP-1) stimulate insulin secretion in a glucose-dependent manner and are thus classified as incretins. The structure of mammalian GIP is well conserved and both the N-terminus and central region of the molecule are important for biological activity. Following secretion, GIP is metabolized by the endoprotease dipeptidyl peptidase IV (DPP-IV). In addition to its insulinotropic activity, GIP exerts a number of additional actions including promotion of growth and survival of the pancreatic beta-cell and stimulation of adipogenesis. The brain, bone, cardiovascular system, and gastrointestinal tract are additional targets of GIP. The GIP receptor is a member of the B-family of G protein-coupled receptors and activation results in the stimulation of adenylyl cyclase and Ca(2+)-independent phospholipase A(2) and activation of protein kinase (PK) A and PKB. The Mek1/2-Erk1/2 and p38 MAP kinase signaling pathways are among the downstream pathways involved in the regulation of beta-cell function. GIP also increases expression of the anti-apoptotic Bcl-2 and decreases expression of the pro-apoptotic Bax, resulting in reduced beta-cell death. In adipose tissue, GIP interacts with insulin to increase lipoprotein lipase activity and lipogenesis. There is significant interest in potential clinical applications for GIP analogs and both agonists and antagonists have been developed for preclinical studies.


Subject(s)
Gastric Inhibitory Polypeptide/genetics , Gastric Inhibitory Polypeptide/metabolism , Amino Acid Sequence , Animals , Gastric Inhibitory Polypeptide/chemistry , Gene Expression Regulation , Humans , Mammals/metabolism
16.
J Biol Chem ; 284(16): 10764-73, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19233842

ABSTRACT

Therapeutics based on the actions of the incretin hormones, glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), have recently been introduced for the treatment of type 2 diabetes mellitus. The serine/threonine kinase Akt is a major mediator of incretin action on the pancreatic islet, increasing beta-cell mass and function and promoting beta-cell survival. The mechanisms underlying incretin activation of Akt are thought to involve an essential phosphoinositide 3-kinase-mediated phosphorylation of threonine 308, similar to the prototypical Akt activator, insulin-like growth factor-I (IGF-I). In this study, using activity assays on immunoprecipitated Akt, we discovered that GIP and GLP-1 were capable of stimulating Akt in the INS-1 beta-cell line and isolated mouse islets via a mechanism that did not require phosphoinositide 3-kinase or phosphorylation of Thr(308) and Ser(473), and this pathway involved the production of cAMP. Furthermore, we found that GIP stimulated anti-apoptotic signaling via this alternate mode of Akt activation. We conclude that incretins can activate Akt via a novel noncanonical mechanism that may provide an alternative therapeutic target for the treatment of type 2 diabetes mellitus and have broader implications for Akt physiology in human health and disease.


Subject(s)
Gastric Inhibitory Polypeptide/metabolism , Incretins/metabolism , Insulin-Secreting Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adenylyl Cyclases/metabolism , Animals , Cell Line , Colforsin/metabolism , Enzyme Activation , Gastric Inhibitory Polypeptide/genetics , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/cytology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Rats
17.
Diabetes ; 58(3): 641-51, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19073764

ABSTRACT

OBJECTIVE: The endopeptidase dipeptidyl peptidase-IV (DPP-IV) has been shown to NH2-terminally truncate incretin hormones, glucose-dependent insulinotropic polypeptide, and glucagon-like peptide-1, thus ablating their ability to potentiate glucose-stimulated insulin secretion. Increasing the circulating levels of incretins through administration of DPP-IV inhibitors has therefore been introduced as a therapeutic approach for the treatment of type 2 diabetes. DPP-IV inhibitor treatment has also been shown to preserve islet mass in rodent models of type 1 diabetes. The current study was initiated to define the effects of the DPP-IV inhibitor sitagliptin (MK0431) on transplanted islet survival in nonobese diabetic (NOD) mice, an autoimmune type 1 diabetes model. RESEARCH DESIGN AND METHODS: Effects of MK0431 on islet graft survival in diabetic NOD mice were determined with metabolic studies and micropositron emission tomography imaging, and its underlying molecular mechanisms were assessed. RESULTS: Treatment of NOD mice with MK0431 before and after islet transplantation resulted in prolongation of islet graft survival, whereas treatment after transplantation alone resulted in small beneficial effects compared with nontreated controls. Subsequent studies demonstrated that MK0431 pretreatment resulted in decreased insulitis in diabetic NOD mice and reduced in vitro migration of isolated splenic CD4+ T-cells. Furthermore, in vitro treatment of splenic CD4+ T-cells with DPP-IV resulted in increased migration and activation of protein kinase A (PKA) and Rac1. CONCLUSIONS: Treatment with MK0431 therefore reduced the effect of autoimmunity on graft survival partially by decreasing the homing of CD4+ T-cells into pancreatic beta-cells through a pathway involving cAMP/PKA/Rac1 activation.


Subject(s)
Diabetes Mellitus, Type 1/surgery , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Graft Survival/drug effects , Graft Survival/immunology , Islets of Langerhans Transplantation/immunology , Pyrazines/pharmacology , T-Lymphocytes/immunology , Triazoles/pharmacology , Animals , Graft Rejection/prevention & control , Male , Mice , Mice, Inbred NOD , Sitagliptin Phosphate , Transplantation, Isogeneic/physiology
18.
Diabetes ; 57(5): 1331-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18299314

ABSTRACT

OBJECTIVE: Dipeptidyl peptidase-IV (DPP-IV) inhibitors have been introduced as therapeutics for type 2 diabetes. They partially act by blocking degradation of the incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), thus increasing circulating levels of active hormones. In addition to their insulinotropic actions, GLP-1 and GIP also promote beta-cell proliferation and survival, and DPP-IV inhibitors exert similar effects in rodent type 2 diabetes models. The study objective was to establish whether DPP-IV inhibitor treatment prolonged survival of transplanted islets and to determine whether positron emission tomography (PET) was appropriate for quantifying the effect of inhibition on islet mass. RESEARCH DESIGN & METHODS: Effects of the DPP-IV inhibitor MK0431 (sitagliptin) on glycemic control and functional islet mass in a streptozotocin (STZ)-induced type 1 diabetes mouse model were determined with metabolic studies and microPET imaging. RESULTS: The type 1 diabetes mouse model exhibited elevated plasma DPP-IV levels that were substantially inhibited in mice on an MK0431 diet. Residual beta-cell mass was extremely low in STZ-induced diabetic mice, and although active GLP-1 levels were increased by the MK0431 diet, there were no significant effects on glycemic control. After islet transplantation, mice fed normal diet rapidly lost their ability to regulate blood glucose, reflecting the suboptimal islet transplant. By contrast, the MK0431 group fully regulated blood glucose throughout the study, and PET imaging demonstrated a profound protective effect of MK0431 on islet graft size. CONCLUSIONS: Treatment with a DPP-IV inhibitor can prolong islet graft retention in an animal model of type 1 diabetes.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Graft Survival/drug effects , Hypoglycemic Agents/pharmacology , Islets of Langerhans Transplantation/immunology , Pyrazines/pharmacology , Triazoles/pharmacology , Animals , Blood Glucose/metabolism , Cell Culture Techniques , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Islets of Langerhans/cytology , Male , Mice , Mice, Inbred C57BL , Sitagliptin Phosphate
19.
Mol Cell Biol ; 28(5): 1644-56, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18086876

ABSTRACT

The cyclic AMP (cAMP)/protein kinase A (PKA) cascade plays a central role in beta-cell proliferation and apoptosis. Here, we show that the incretin hormone glucose-dependent insulinotropic polypeptide (GIP) stimulates expression of the antiapoptotic Bcl-2 gene in pancreatic beta cells through a pathway involving AMP-activated protein kinase (AMPK), cAMP-responsive CREB coactivator 2 (TORC2), and cAMP response element binding protein (CREB). Stimulation of beta-INS-1 (clone 832/13) cells with GIP resulted in increased Bcl-2 promoter activity. Analysis of the rat Bcl-2 promoter revealed two potential cAMP response elements, one of which (CRE-I [GTGACGTAC]) was shown, using mutagenesis and deletion analysis, to be functional. Subsequent studies established that GIP increased the nuclear localization of TORC2 and phosphorylation of CREB serine 133 through a pathway involving PKA activation and reduced AMPK phosphorylation. At the nuclear level, phospho-CREB and TORC2 were demonstrated to bind to CRE-I of the Bcl-2 promoter, and GIP treatment resulted in increases in their interaction. Furthermore, GIP-mediated cytoprotection was partially reversed by small interfering RNA-mediated reduction in BCL-2 or TORC2/CREB or by pharmacological activation of AMPK. The antiapoptotic effect of GIP in beta cells is therefore partially mediated through a novel mode of transcriptional regulation of Bcl-2 involving cAMP/PKA/AMPK-dependent regulation of CREB/TORC2 activity.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Insulin-Secreting Cells/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription Factors/metabolism , Up-Regulation/drug effects , Animals , Caspase 3/analysis , Caspase 3/metabolism , Cell Culture Techniques , Cell Nucleus/metabolism , Cell Survival/drug effects , Cells, Cultured , Clone Cells/metabolism , Culture Media , Cyclic AMP Response Element-Binding Protein/genetics , Dose-Response Relationship, Drug , Gastric Inhibitory Polypeptide/metabolism , Gene Expression , Genes, Reporter , Luciferases/analysis , Luciferases/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Interference , Rats , Statistics as Topic , Trans-Activators , Transcription Factors/genetics , Transfection , beta-Galactosidase/metabolism
20.
Can J Diabetes ; 32(2): 131-9, 2008.
Article in English | MEDLINE | ID: mdl-27056542

ABSTRACT

There is a growing interest in developing therapeutic strategies for type 2 diabetes based on the actions of the hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP). These hormones are the major incretins released from the intestine in response to nutrient ingestion, and they stimulate insulin secretion in a glucosedependent manner. Both peptides are degraded by the enzyme dipeptidyl peptidase-4 (DPP-4), thus terminating their actions. Studies in animal models of diabetes have shown that the incretins also exert a number of additional actions that improve glucose disposal. GLP-1 reduces food intake and gastric emptying, as well as inhibiting glucagon secretion. Injectable formulations of DPP-4-resistant GLP-1-related peptides (incretin mimetics) that are now in clinical use (exenatide) or undergoing trials (e.g. liraglutide) have been shown to reduce fasting and postprandial glucose and glycosylated hemoglobin (A1C) levels and induce weight loss. Oral administration of DPP-4 inhibitors potentiates the actions of incretins released during a meal. Clinical trials have demonstrated that DPP-4 inhibitors are weight-neutral drugs that also effectively reduce plasma glucose and A1C levels. One inhibitor, sitagliptin, is now available in Canada and the United States, and another, vildagliptin, has recently been approved by the European Union. Other inhibitors are under development. Preclinical studies indicate that treatment with incretin mimetics or DPP-4 inhibitors also preserves beta cell mass by exerting mitogenetic and prosurvival effects. It is not known whether similar effects occur in humans.

SELECTION OF CITATIONS
SEARCH DETAIL
...