Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Biol Chem ; 288(34): 24825-33, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23853095

ABSTRACT

In pancreatic ß-cells, glucose induces the binding of the transcription factor pancreatic duodenal homeobox-1 (PDX-1) to the insulin gene promoter to activate insulin gene transcription. At low glucose levels, glycogen synthase kinase 3ß (GSK3ß) is known to phosphorylate PDX-1 on C-terminal serine residues, which triggers PDX-1 proteasomal degradation. We previously showed that the serine/threonine Per-Arnt-Sim domain-containing kinase (PASK) regulates insulin gene transcription via PDX-1. However, the mechanisms underlying this regulation are unknown. In this study, we aimed to identify the role of PASK in the regulation of PDX-1 phosphorylation, protein expression, and stability in insulin-secreting cells and isolated rodent islets of Langerhans. We observed that glucose induces a decrease in overall PDX-1 serine phosphorylation and that overexpression of WT PASK mimics this effect. In vitro, PASK directly phosphorylates GSK3ß on its inactivating phosphorylation site Ser(9). Overexpression of a kinase-dead (KD), dominant negative version of PASK blocks glucose-induced Ser(9) phosphorylation of GSK3ß. Accordingly, GSK3ß Ser(9) phosphorylation is reduced in islets from pask-null mice. Overexpression of WT PASK or KD GSK3ß protects PDX-1 from degradation and results in increased PDX-1 protein abundance. Conversely, overexpression of KD PASK blocks glucose-induction of PDX-1 protein. We conclude that PASK phosphorylates and inactivates GSK3ß, thereby preventing PDX-1 serine phosphorylation and alleviating GSK3ß-mediated PDX-1 protein degradation in pancreatic ß-cells.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/metabolism , Animals , Glucose/pharmacology , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Hep G2 Cells , Homeodomain Proteins/genetics , Humans , Insulin-Secreting Cells/cytology , Male , Mice , Mutation , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Protein Stability/drug effects , Rats , Rats, Wistar , Sweetening Agents/pharmacology , Trans-Activators/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
2.
bioRxiv ; 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38328157

ABSTRACT

Large library docking can reveal unexpected chemotypes that complement the structures of biological targets. Seeking new agonists for the cannabinoid-1 receptor (CB1R), we docked 74 million tangible molecules, prioritizing 46 high ranking ones for de novo synthesis and testing. Nine were active by radioligand competition, a 20% hit-rate. Structure-based optimization of one of the most potent of these (Ki = 0.7 uM) led to '4042, a 1.9 nM ligand and a full CB1R agonist. A cryo-EM structure of the purified enantiomer of '4042 ('1350) in complex with CB1R-Gi1 confirmed its docked pose. The new agonist was strongly analgesic, with generally a 5-10-fold therapeutic window over sedation and catalepsy and no observable conditioned place preference. These findings suggest that new cannabinoid chemotypes may disentangle characteristic cannabinoid side-effects from their analgesia, supporting the further development of cannabinoids as pain therapeutics.

3.
Nat Commun ; 14(1): 3953, 2023 07 04.
Article in English | MEDLINE | ID: mdl-37402735

ABSTRACT

Urocortin 2 (UCN2) acts as a ligand for the G protein-coupled receptor corticotropin-releasing hormone receptor 2 (CRHR2). UCN2 has been reported to improve or worsen insulin sensitivity and glucose tolerance in vivo. Here we show that acute dosing of UCN2 induces systemic insulin resistance in male mice and skeletal muscle. Inversely, chronic elevation of UCN2 by injection with adenovirus encoding UCN2 resolves metabolic complications, improving glucose tolerance. CRHR2 recruits Gs in response to low concentrations of UCN2, as well as Gi and ß-Arrestin at high concentrations of UCN2. Pre-treating cells and skeletal muscle ex vivo with UCN2 leads to internalization of CRHR2, dampened ligand-dependent increases in cAMP, and blunted reductions in insulin signaling. These results provide mechanistic insights into how UCN2 regulates insulin sensitivity and glucose metabolism in skeletal muscle and in vivo. Importantly, a working model was derived from these results that unifies the contradictory metabolic effects of UCN2.


Subject(s)
Insulin Resistance , Animals , Male , Mice , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Glucose/metabolism , Insulin , Ligands , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Urocortins/genetics , Urocortins/metabolism
4.
J Biol Chem ; 286(51): 44005-44014, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22065581

ABSTRACT

PAS kinase (PASK) is a glucose-regulated protein kinase involved in the control of pancreatic islet hormone release and insulin sensitivity. We aimed here to identify mutations in the PASK gene that may be associated with young-onset diabetes in humans. We screened 18 diabetic probands with unelucidated maturity-onset diabetes of the young (MODY). We identified two rare nonsynonymous mutations in the PASK gene (p.L1051V and p.G1117E), each of which was found in a single MODY family. Wild type or mutant PASKs were expressed in HEK 293 cells. Kinase activity of the affinity-purified proteins was assayed as autophosphorylation at amino acid Thr307 or against an Ugp1p-derived peptide. Whereas the PASK p.G1117E mutant displayed a ∼25% increase with respect to wild type PASK in the extent of autophosphorylation, and a ∼2-fold increase in kinase activity toward exogenous substrates, the activity of the p.L1051V mutant was unchanged. Amino acid Gly1117 is located in an α helical region opposing the active site of PASK and may elicit either: (a) a conformational change that increases catalytic efficiency or (b) a diminished inhibitory interaction with the PAS domain. Mouse islets were therefore infected with adenoviruses expressing wild type or mutant PASK and the regulation of insulin secretion was examined. PASK p.G1117E-infected islets displayed a 4-fold decrease in glucose-stimulated (16.7 versus 3 mM) insulin secretion, chiefly reflecting a 4.5-fold increase in insulin release at low glucose. In summary, we have characterized a rare mutation (p.G1117E) in the PASK gene from a young-onset diabetes family, which modulates glucose-stimulated insulin secretion.


Subject(s)
Glucose/metabolism , Insulin/metabolism , Islets of Langerhans/cytology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Adult , Animals , Cell Line , Diabetes Mellitus/metabolism , Genomics , Glucagon/metabolism , HEK293 Cells , HSP70 Heat-Shock Proteins/metabolism , Humans , Insulin Secretion , Male , Membrane Proteins/metabolism , Models, Genetic , Mutagenesis , Phosphorylation , Rats , Rats, Wistar , Recombinant Proteins/metabolism
5.
Biochim Biophys Acta ; 1801(3): 289-98, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19715772

ABSTRACT

The concept of glucolipotoxicity refers to the combined, deleterious effects of elevated glucose and fatty acid levels on pancreatic beta-cell function and survival. Significant progress has been made in recent years towards a better understanding of the cellular and molecular basis of glucolipotoxicity in the beta cell. The permissive effect of elevated glucose on the detrimental actions of fatty acids stems from the influence of glucose on intracellular fatty acid metabolism, promoting the synthesis of cellular lipids. The combination of excessive levels of fatty acids and glucose therefore leads to decreased insulin secretion, impaired insulin gene expression, and beta-cell death by apoptosis, all of which probably have distinct underlying mechanisms. Recent studies from our laboratory have identified several pathways implicated in fatty acid inhibition of insulin gene expression, including the extracellular-regulated kinase (ERK1/2) pathway, the metabolic sensor Per-Arnt-Sim kinase (PASK), and the ATF6 branch of the unfolded protein response. We have also confirmed in vivo in rats that the decrease in insulin gene expression is an early defect which precedes any detectable abnormality in insulin secretion. While the role of glucolipotoxicity in humans is still debated, the inhibitory effects of chronically elevated fatty acid levels has been clearly demonstrated in several studies, at least in individuals genetically predisposed to developing type 2 diabetes. It is therefore likely that glucolipotoxicity contributes to beta-cell failure in type 2 diabetes as well as to the decline in beta-cell function observed after the onset of the disease.


Subject(s)
Fatty Acids/metabolism , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Lipid Metabolism Disorders/metabolism , Animals , Female , Humans , Insulin Resistance , Male , Obesity/metabolism
6.
Chem Sci ; 12(33): 10990-11003, 2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34522296

ABSTRACT

Brain functions rely on neurotransmitters that mediate communication between billions of neurons. Disruption of this communication can result in a plethora of psychiatric and neurological disorders. In this work, we combine molecular dynamics simulations, live-cell biosensor and electrophysiological assays to investigate the action of the neurotransmitter dopamine at the dopaminergic D2 receptor (D2R). The study of dopamine and closely related chemical probes reveals how neurotransmitter binding translates into the activation of distinct subsets of D2R effectors (i.e.: Gi2, GoB, Gz and ß-arrestin 2). Ligand interactions with key residues in TM5 (S5.42) and TM6 (H6.55) in the D2R binding pocket yield a dopamine-like coupling signature, whereas exclusive TM5 interaction is typically linked to preferential G protein coupling (in particular GoB) over ß-arrestin. Further experiments for serotonin receptors indicate that the reported molecular mechanism is shared by other monoaminergic neurotransmitter receptors. Ultimately, our study highlights how sequence variation in position 6.55 is used by nature to fine-tune ß-arrestin recruitment and in turn receptor signaling and internalization of neurotransmitter receptors.

7.
Mol Brain ; 14(1): 151, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34583741

ABSTRACT

The atypical chemokine receptor 3, ACKR3, is a G protein-coupled receptor, which does not couple to G proteins but recruits ßarrestins. At present, ACKR3 is considered a target for cancer and cardiovascular disorders, but less is known about the potential of ACKR3 as a target for brain disease. Further, mouse lines have been created to identify cells expressing the receptor, but there is no tool to visualize and study the receptor itself under physiological conditions. Here, we engineered a knock-in (KI) mouse expressing a functional ACKR3-Venus fusion protein to directly detect the receptor, particularly in the adult brain. In HEK-293 cells, native and fused receptors showed similar membrane expression, ligand induced trafficking and signaling profiles, indicating that the Venus fusion does not alter receptor signaling. We also found that ACKR3-Venus enables direct real-time monitoring of receptor trafficking using resonance energy transfer. In ACKR3-Venus knock-in mice, we found normal ACKR3 mRNA levels in the brain, suggesting intact gene transcription. We fully mapped receptor expression across 14 peripheral organs and 112 brain areas and found that ACKR3 is primarily localized to the vasculature in these tissues. In the periphery, receptor distribution aligns with previous reports. In the brain there is notable ACKR3 expression in endothelial vascular cells, hippocampal GABAergic interneurons and neuroblast neighboring cells. In conclusion, we have generated Ackr3-Venus knock-in mice with a traceable ACKR3 receptor, which will be a useful tool to the research community for interrogations about ACKR3 biology and related diseases.


Subject(s)
Bacterial Proteins/genetics , Brain/blood supply , Gene Knock-In Techniques , Genes, Reporter , Luminescent Proteins/genetics , Receptors, CXCR/genetics , Animals , Bacterial Proteins/analysis , Bacterial Proteins/pharmacokinetics , Biomarkers , Computer Systems , Endothelial Cells/chemistry , Endothelial Cells/cytology , GABAergic Neurons/chemistry , GABAergic Neurons/cytology , HEK293 Cells , Humans , Interneurons/chemistry , Interneurons/cytology , Ligands , Luminescent Proteins/analysis , Luminescent Proteins/pharmacokinetics , Mice , Organ Specificity , Receptors, CXCR/analysis , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacokinetics , Tissue Distribution , beta-Arrestin 1/metabolism
8.
Commun Biol ; 3(1): 719, 2020 11 27.
Article in English | MEDLINE | ID: mdl-33247181

ABSTRACT

Protease-activated receptor-2 (PAR2) is involved in inflammatory responses and pain, therefore representing a promising therapeutic target for the treatment of immune-mediated inflammatory diseases. However, as for other GPCRs, PAR2 can activate multiple signaling pathways and those involved in inflammatory responses remain poorly defined. Here, we describe a new selective and potent PAR2 inhibitor (I-287) that shows functional selectivity by acting as a negative allosteric regulator on Gαq and Gα12/13 activity and their downstream effectors, while having no effect on Gi/o signaling and ßarrestin2 engagement. Such selective inhibition of only a subset of the pathways engaged by PAR2 was found to be sufficient to block inflammation in vivo. In addition to unraveling the PAR2 signaling pathways involved in the pro-inflammatory response, our study opens the path toward the development of new functionally selective drugs with reduced liabilities that could arise from blocking all the signaling activities controlled by the receptor.


Subject(s)
Anti-Inflammatory Agents/pharmacology , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Receptor, PAR-2/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Bioluminescence Resonance Energy Transfer Techniques , Cell Line, Tumor , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Interleukin-8/metabolism , Male , Mice , Mice, Inbred C57BL , beta-Arrestins/metabolism
9.
iScience ; 14: 47-57, 2019 Apr 26.
Article in English | MEDLINE | ID: mdl-30925410

ABSTRACT

G protein-coupled receptors are key signaling molecules and major targets for pharmaceuticals. The concept of ligand-dependent biased signaling raises the possibility of developing drugs with improved efficacy and safety profiles, yet translating this concept to native tissues remains a major challenge. Whether drug activity profiling in recombinant cell-based assays, traditionally used for drug discovery, has any relevance to physiology is unknown. Here we focused on the mu opioid receptor, the unrivalled target for pain treatment and also the key driver for the current opioid crisis. We selected a set of clinical and novel mu agonists, and profiled their activities in transfected cell assays using advanced biosensors and in native neurons from knock-in mice expressing traceable receptors endogenously. Our data identify Gi-biased agonists, including buprenorphine, and further show highly correlated drug activities in the two otherwise very distinct experimental systems, supporting in vivo translatability of biased signaling for mu opioid drugs.

10.
Mol Genet Metab ; 95(3): 117-26, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18762440

ABSTRACT

Fat cell lipolysis, the cleavage of triglycerides and release of fatty acids and glycerol, evolved to enable survival during prolonged food deprivation but is paradoxically increased in obesity, in which a surfeit of all energy metabolites is found. Essential, previously-unsuspected components have been discovered in the lipolytic machinery, at the protective interface of the lipid droplet surface and in the signaling pathways that control lipolysis. At least two adipocyte lipases are important for controlling lipolysis, hormone-sensitive lipase (HSL) and adipocyte triglyceride lipase (ATGL). Perilipin (PLIN) and possibly other proteins of the lipid droplet surface are master regulators of lipolysis, protecting or exposing the triglyceride core of the droplet to lipases. The prototypes for hormonal lipolytic control are beta adrenergic stimulation and suppression by insulin, both of which affect cyclic AMP levels and hence the protein kinase A-mediated phosphorylation of HSL and PLIN. Newly-recognized mediators of lipolysis include atrial natriuretic peptide, cyclic GMP, the ketone body 3-hydroxybutyrate, AMP kinase and mitogen-activated kinases. Lipolysis must be interpreted in its physiological context since similar rates of basal or stimulated lipolysis occur under different conditions and by different mechanisms. Age, sex, anatomical site, genotype and species differences are each important variables. Manipulation of lipolysis has therapeutic potential in several inborn errors and in the metabolic syndrome that frequently complicates obesity.


Subject(s)
Energy Metabolism , Lipolysis , Signal Transduction , Adipocytes/enzymology , Adipocytes/metabolism , Animals , Gene Expression Regulation , Humans , Lipase/genetics , Lipase/metabolism , Obesity/metabolism , Obesity/therapy
11.
Brain Struct Funct ; 223(3): 1275-1296, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29110094

ABSTRACT

GPR88 is an orphan G-protein coupled receptor originally characterized as a striatal-enriched transcript and is a potential target for neuropsychiatric disorders. At present, gene knockout studies in the mouse have essentially focused on striatal-related functions and a comprehensive knowledge of GPR88 protein distribution and function in the brain is still lacking. Here, we first created Gpr88-Venus knock-in mice expressing a functional fluorescent receptor to fine-map GPR88 localization in the brain. The receptor protein was detected in neuronal soma, fibers and primary cilia depending on the brain region, and remarkably, whole-brain mapping revealed a yet unreported layer-4 cortical lamination pattern specifically in sensory processing areas. The unique GPR88 barrel pattern in L4 of the somatosensory cortex appeared 3 days after birth and persisted into adulthood, suggesting a potential function for GPR88 in sensory integration. We next examined Gpr88 knockout mice for cortical structure and behavioral responses in sensory tasks. Magnetic resonance imaging of live mice revealed abnormally high fractional anisotropy, predominant in somatosensory cortex and caudate putamen, indicating significant microstructural alterations in these GPR88-enriched areas. Further, behavioral analysis showed delayed responses in somatosensory-, visual- and olfactory-dependent tasks, demonstrating a role for GPR88 in the integration rather than perception of sensory stimuli. In conclusion, our data show for the first time a prominent role for GPR88 in multisensory processing. Because sensory integration is disrupted in many psychiatric diseases, our study definitely positions GPR88 as a target to treat mental disorders perhaps via activity on cortical sensory networks.


Subject(s)
Bacterial Proteins/metabolism , Brain Mapping , Brain/metabolism , Luminescent Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , ADP-Ribosylation Factors/metabolism , Animals , Bacterial Proteins/genetics , Brain/cytology , Brain/diagnostic imaging , Carrier Proteins/metabolism , Cells, Cultured , Discrimination, Psychological/physiology , Endodeoxyribonucleases , Female , G-Protein-Coupled Receptor Kinase 2/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , HEK293 Cells , Humans , Luminescent Proteins/genetics , Magnetic Resonance Imaging , Male , Mice , Mice, Transgenic , Nuclear Proteins/metabolism , Odorants , Phosphopyruvate Hydratase/metabolism , Psychomotor Performance/physiology , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Recognition, Psychology/physiology , Transfection
12.
Islets ; 6(4): e982376, 2014.
Article in English | MEDLINE | ID: mdl-25437380

ABSTRACT

The transcription factor Pancreatic and Duodenal Homeobox-1 (PDX-1) plays a major role in the development and function of pancreatic ß-cells and its mutation results in diabetes. In adult ß-cells, glucose stimulates transcription of the insulin gene in part by regulating PDX-1 expression, stability and activity. Glucose is also thought to modulate PDX-1 nuclear translocation but in vitro studies examining nucleo-cytoplasmic shuttling of endogenous or ectopically expressed PDX-1 in insulin-secreting cell lines have led to conflicting results. Here we show that endogenous PDX-1 undergoes translocation from the cytoplasm to the nucleus in response to glucose in dispersed rat islets but not in insulin-secreting MIN6, HIT-T15, or INS832/13 cells. Interestingly, however, we found that a PDX-1-GFP fusion protein can shuttle from the cytoplasm to the nucleus in response to glucose stimulation in HIT-T15 cells. Our results suggest that the regulation of endogenous PDX-1 sub-cellular localization by glucose is observed in primary islets and that care should be taken when interpreting data from insulin-secreting cell lines.


Subject(s)
Glucose/pharmacology , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Trans-Activators/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Immunohistochemistry , Male , Rats , Rats, Wistar
13.
PLoS One ; 7(4): e36200, 2012.
Article in English | MEDLINE | ID: mdl-22558381

ABSTRACT

BACKGROUND: Type 2 diabetes is characterized by pancreatic ß-cell dysfunction and is associated with low-grade inflammation. Recent observations suggest that the signalling cascade activated by lipopolysaccharides (LPS) binding to Toll-Like Receptor 4 (TLR4) exerts deleterious effects on pancreatic ß-cell function; however, the molecular mechanisms of these effects are incompletely understood. In this study, we tested the hypothesis that LPS alters insulin gene expression via TLR4 and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in islets. METHODOLOGY/PRINCIPAL FINDINGS: A 24-h exposure of isolated human, rat and mouse islets of Langerhans to LPS dose-dependently reduced insulin gene expression. This was associated in mouse and rat islets with decreased mRNA expression of pancreas-duodenum homebox-1 (PDX-1) and mammalian homologue of avian MafA/l-Maf (MafA). Accordingly, LPS exposure also decreased glucose-induced insulin secretion. LPS repression of insulin, PDX-1 and MafA expression, as well as its inhibition of insulin secretion, were not observed in islets from TLR4-deficient mice. LPS inhibition of ß-cell gene expression in rat islets was prevented by inhibition of the NF-κB pathway, but not the p38 mitogen-activated protein kinase (p38 MAPK) pathway. CONCLUSIONS/SIGNIFICANCE: Our findings demonstrate that LPS inhibit ß-cell gene expression in a TLR4-dependent manner and via NF-κB signaling in pancreatic islets, suggesting a novel mechanism by which the gut microbiota might affect pancreatic ß-cell function.


Subject(s)
Gene Expression Regulation/drug effects , Insulin/genetics , Islets of Langerhans/drug effects , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Cell Line , Homeodomain Proteins/metabolism , Humans , In Vitro Techniques , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Male , Mice , NF-kappa B/antagonists & inhibitors , RNA Precursors/genetics , RNA Precursors/metabolism , Rats , Toll-Like Receptor 4/deficiency , Trans-Activators/metabolism
14.
Diabetes Res Clin Pract ; 87(3): 322-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20092903

ABSTRACT

UNLABELLED: Prolonged exposure of pancreatic beta-cells to elevated levels of glucose and fatty acids adversely affects insulin secretion and gene expression. AIM: To examine whether the GLP-1 agonist exenatide or the inhibitor of the GLP-1-degrading enzyme dipeptidyl peptidase 4 (DPP-4) sitagliptin rescue insulin gene expression in rats infused for 72h with glucose+Intralipid, independently from their glucose-lowering action. METHODS: Wistar rats were infused alternatively with glucose or Intralipid for cycles of 4h each for a total of 72h. The animals received exenatide (5microg/kg/day IV) or sitagliptin (5mg/kg/day IV) continuously starting 4 days prior to and continuing throughout the 3-day infusion period. RESULTS: Plasma glucose, fatty acids, insulin and C-peptide levels were unaffected by exenatide or sitagliptin treatment during the infusion period. Insulin mRNA levels increased in response to the glucose infusion, but this increase was abolished in islets from rats receiving glucose+Intralipid. Neither exenatide nor sitagliptin administration rescued insulin mRNA in glucose+Intralipid infused rats. CONCLUSIONS: Neither a GLP-1 agonist nor a DPP-4 inhibitor, at doses that do not alter blood glucose levels, prevented the inhibition of insulin gene expression in this in vivo model of glucolipotoxicity.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors , Gene Expression/drug effects , Glucagon-Like Peptide 1/agonists , Hyperglycemia/genetics , Hyperlipidemias/genetics , Insulin-Secreting Cells/drug effects , Insulin/genetics , Analysis of Variance , Animals , Blood Glucose/metabolism , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Eating/drug effects , Exenatide , Fat Emulsions, Intravenous , Glucagon-Like Peptide 1/metabolism , Glucose , Hyperglycemia/chemically induced , Hyperglycemia/metabolism , Hyperlipidemias/chemically induced , Hyperlipidemias/metabolism , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Lipids/blood , Male , Peptides/pharmacology , Pyrazines/pharmacology , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sitagliptin Phosphate , Triazoles/pharmacology , Venoms/pharmacology
15.
Diabetes ; 58(9): 2048-58, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19502418

ABSTRACT

OBJECTIVE: Prolonged exposure of pancreatic beta-cells to simultaneously elevated levels of fatty acids and glucose (glucolipotoxicity) impairs insulin gene transcription. However, the intracellular signaling pathways mediating these effects are mostly unknown. This study aimed to ascertain the role of extracellular-regulated kinases (ERKs)1/2, protein kinase B (PKB), and Per-Arnt-Sim kinase (PASK) in palmitate inhibition of insulin gene expression in pancreatic beta-cells. RESEARCH DESIGN AND METHODS: MIN6 cells and isolated rat islets were cultured in the presence of elevated glucose, with or without palmitate or ceramide. ERK1/2 phosphorylation, PKB phosphorylation, and PASK expression were examined by immunoblotting and real-time PCR. The role of these kinases in insulin gene expression was assessed using pharmacological and molecular approaches. RESULTS: Exposure of MIN6 cells and islets to elevated glucose induced ERK1/2 and PKB phosphorylation, which was further enhanced by palmitate. Inhibition of ERK1/2, but not of PKB, partially prevented the inhibition of insulin gene expression in the presence of palmitate or ceramide. Glucose-induced expression of PASK mRNA and protein levels was reduced in the presence of palmitate. Overexpression of wild-type PASK increased insulin and pancreatic duodenal homeobox-1 gene expression in MIN6 cells and rat islets incubated with glucose and palmitate, whereas overexpression of a kinase-dead PASK mutant in rat islets decreased expression of insulin and pancreatic duodenal homeobox-1 and increased C/EBPbeta expression. CONCLUSIONS: Both the PASK and ERK1/2 signaling pathways mediate palmitate inhibition of insulin gene expression. These findings identify PASK as a novel mediator of glucolipotoxicity on the insulin gene in pancreatic beta-cells.


Subject(s)
Insulin-Secreting Cells/physiology , Insulin/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Line , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Glucaric Acid/pharmacology , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/physiology , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Palmitates/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Proto-Oncogene Proteins c-akt/metabolism , RNA Precursors/metabolism , RNA, Messenger/metabolism , Rats , Sphingosine/analogs & derivatives , Sphingosine/pharmacology
16.
Diabetes ; 57(9): 2432-7, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18559658

ABSTRACT

OBJECTIVE: The G-protein-coupled receptor GPR40 is expressed in pancreatic beta-cells and is activated by long-chain fatty acids. Gene deletion studies have shown that GPR40 mediates, at least in part, fatty acid-amplification of glucose-induced insulin secretion (GSIS) but is not implicated in GSIS itself. However, the role of GPR40 in the long-term effects of fatty acids on insulin secretion remains controversial. This study aimed to test the hypothesis that GPR40 plays a role in insulin secretion after high-fat feeding. RESEARCH DESIGN AND METHOD GPR40 knockout (KO) mice on a C57BL/6 background and their wild-type (WT) littermates were fed a high-fat diet (HFD) for 11 weeks. Glucose tolerance, insulin tolerance, and insulin secretion in response to glucose and Intralipid were assessed during the course of the diet period. RESULTS: GPR40 KO mice had fasting hyperglycemia. They became as obese, glucose intolerant, and insulin resistant as their WT littermates given HFD and developed a similar degree of liver steatosis. Their fasting blood glucose levels increased earlier than those of control mice during the course of the HFD. The remarkable increase in insulin secretory responses to intravenous glucose and Intralipid seen in WT mice after HFD was of much lower magnitude in GPR40 KO mice. CONCLUSIONS: GPR40 plays a role not only in fatty acid modulation of insulin secretion, but also in GSIS after high-fat feeding. These observations raise doubts on the validity of a therapeutic approach based on GPR40 antagonism for the treatment of type 2 diabetes.


Subject(s)
Dietary Fats/pharmacology , Hyperglycemia/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Blood Glucose/metabolism , Eating/physiology , Fatty Acids/metabolism , Gene Expression/physiology , Glucose Intolerance/metabolism , Glucose Intolerance/physiopathology , Hyperglycemia/physiopathology , Insulin Secretion , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Obesity/physiopathology , Receptors, G-Protein-Coupled/genetics
17.
Diabetes ; 57(2): 424-31, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17991758

ABSTRACT

OBJECTIVE: Prolonged exposure of isolated islets of Langerhans to elevated levels of fatty acids, in the presence of high glucose, impairs insulin gene expression via a transcriptional mechanism involving nuclear exclusion of pancreas-duodenum homeobox-1 (Pdx-1) and loss of MafA expression. Whether such a phenomenon also occurs in vivo is unknown. Our objective was therefore to ascertain whether chronic nutrient oversupply inhibits insulin gene expression in vivo. RESEARCH DESIGN AND METHODS: Wistar rats received alternating 4-h infusions of glucose and Intralipid for a total of 72 h. Control groups received alternating infusions of glucose and saline, saline and Intralipid, or saline only. Insulin and C-peptide secretion were measured under hyperglycemic clamps. Insulin secretion and gene expression were assessed in isolated islets, and beta-cell mass was quantified by morphometric analysis. RESULTS: Neither C-peptide secretion nor insulin sensitivity was different among infusion regimens. Insulin content and insulin mRNA levels were lower in islets isolated from rats infused with glucose plus Intralipid. This was associated with reduced Pdx-1 binding to the endogenous insulin promoter, and an increased proportion of Pdx-1 localized in the cytoplasm versus the nucleus. In contrast, MafA mRNA and protein levels and beta-cell mass and proliferation were unchanged. CONCLUSIONS: Cyclical and alternating infusions of glucose and Intralipid in normal rats inhibit insulin gene expression without affecting insulin secretion or beta-cell mass. We conclude that fatty acid inhibition of insulin gene expression, in the presence of high glucose, is an early functional defect that may contribute to beta-cell failure in type 2 diabetes.


Subject(s)
Fat Emulsions, Intravenous/pharmacology , Gene Expression Regulation/drug effects , Glucose/pharmacology , Homeodomain Proteins/metabolism , Insulin/genetics , Trans-Activators/metabolism , Animals , Blood Glucose/metabolism , C-Peptide/drug effects , C-Peptide/metabolism , Fat Emulsions, Intravenous/administration & dosage , Fatty Acids, Nonesterified/blood , Glucose/administration & dosage , Glucose Tolerance Test , Homeodomain Proteins/drug effects , Hyperglycemia , Infusions, Intravenous , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , RNA, Messenger/genetics , Rats , Rats, Wistar , Trans-Activators/drug effects
18.
J Biol Chem ; 279(39): 40683-9, 2004 Sep 24.
Article in English | MEDLINE | ID: mdl-15220344

ABSTRACT

Hydrolysis of triglycerides is central to energy homeostasis in white adipose tissue (WAT). Hormone-sensitive lipase (HSL) was previously felt to mediate all lipolysis in WAT. Surprisingly, HSL-deficient mice show active HSL-independent lipolysis, suggesting that other lipase(s) also mediate triglyceride hydrolysis. To clarify this, we used functional proteomics to detect non-HSL lipase(s) in mouse WAT. After cell fractionation of intraabdominal WAT, most non-HSL neutral lipase activity is localized in the 100,000 x g infranatant and fat cake fractions. By oleic acid-linked agarose chromatography of infranatant followed by elution in a 3-[(3-cholamidopropyl)dimethylammonio]-1-propanesulfonic acid gradient, we identified two peaks of esterase activity using p-nitrophenyl butyrate as a substrate. One of the peaks contained most of the lipase activity. In the corresponding fractions, gel permeation chromatography and SDS-PAGE, followed by tandem mass spectrometric analysis of excised Coomassie Blue-stained peptides, revealed carboxylesterase 3 (triacylglycerol hydrolase (TGH); EC 3.1.1.1). TGH is also the principle lipase of WAT fat cake extracts. Partially purified WAT TGH had lipase activity as well as lesser but detectable neutral cholesteryl ester hydrolase activity. Western blotting of subcellular fractions of WAT and confocal microscopy of fibroblasts following in vitro adipocytic differentiation are consistent with a distribution of TGH to endoplasmic reticulum, cytosol, and the lipid droplet. TGH is responsible for a major part of non-HSL lipase activity in WAT in vitro and may mediate some or all HSL-independent lipolysis in adipocytes.


Subject(s)
Adipocytes/enzymology , Lipase/physiology , Adipocytes/metabolism , Alkanesulfonic Acids/chemistry , Animals , Blotting, Western , Carboxylesterase/metabolism , Cholesterol/chemistry , Chromatography , Chromatography, Agarose , Chromatography, Gel , Cytosol/metabolism , Electrophoresis, Polyacrylamide Gel , Endoplasmic Reticulum/metabolism , Esterases/metabolism , Fibroblasts/metabolism , Hydrolysis , Lipase/metabolism , Lipid Metabolism , Mass Spectrometry , Mice , Mice, Inbred C57BL , Microscopy, Confocal , NIH 3T3 Cells , Oleic Acid/chemistry , Peptides/chemistry , Sterol Esterase/metabolism , Subcellular Fractions/metabolism , Substrate Specificity , Time Factors , Triglycerides/chemistry
19.
Am J Physiol Endocrinol Metab ; 287(2): E282-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15271647

ABSTRACT

In white adipose tissue, lipolysis can occur by hormone-sensitive lipase (HSL)-dependent or HSL-independent pathways. To study HSL-independent lipolysis, we placed HSL-deficient mice in conditions of increased fatty acid flux: beta-adrenergic stimulation, fasting, and dietary fat loading. Intraperitoneal administration of the beta(3)-adrenergic agonist CL-316243 caused a greater increase in nonesterified fatty acid level in controls (0.33 +/- 0.05 mmol/l) than in HSL(-/-) mice (0.12 +/- 0.01 mmol/l, P < 0.01). Similarly, in isolated adipocytes, lipolytic response to CL-316243 was greatly reduced in HSL(-/-) mice compared with controls. Fasting for

Subject(s)
Adipocytes/metabolism , Adrenergic beta-Agonists/pharmacology , Fasting/metabolism , Fatty Acids/metabolism , Lipolysis/physiology , Sterol Esterase/physiology , Adipocytes/cytology , Adipocytes/drug effects , Analysis of Variance , Animals , Cells, Cultured , Dietary Fats/metabolism , Dietary Fats/pharmacology , Dioxoles/pharmacology , Energy Metabolism/drug effects , Energy Metabolism/genetics , Fatty Acids/pharmacology , Female , Homeostasis/drug effects , Homeostasis/genetics , Lipolysis/drug effects , Male , Mice , Mice, Knockout , Motor Activity/drug effects , Motor Activity/genetics , Sterol Esterase/deficiency , Sterol Esterase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL