Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
2.
Sci Rep ; 6: 29967, 2016 07 18.
Article in English | MEDLINE | ID: mdl-27426254

ABSTRACT

Neurological abnormalities, such as Parkinson-like disorders (PlD), are often co-morbidities of Type 2 Diabetic (T2D) patients, although the epidemiological link between these two disorders remains controversial. The PED/PEA-15 protein represents a possible candidate linking T2D and PD, because it is increased in subjects with T2D and is highly expressed in the brain. To test this hypothesis, we have analyzed the neurological and neurochemical phenotype of transgenic mice overexpressing PED/PEA-15 (tgPED). These mice develop impaired glucose tolerance and insulin resistance, accompanied by neurological features resembling PlD: feet clasping, slow and delayed locomotor movements in different behavioral tests in absence of clear cognitive deficits, ataxia or anxiety. Morphological analysis of the brains showed selective modifications of metabolic activity in the striatal region. In the same region, we have observed 26% decrease of dopamine fibers, confirmed by immunohistochemistry and Western Blot for tyrosine hydroxylase. Moreover, they also showed 48% reduction of dopamine levels in the striatum. Thus the tgPED mice may represent a genetic animal model of neurological disease linked to T2D.


Subject(s)
Insulin Resistance , Parkinson Disease/pathology , Phosphoproteins/metabolism , Animals , Apoptosis Regulatory Proteins , Behavior, Animal , Dopamine/metabolism , Male , Memory , Mice, Transgenic , Motor Activity , Neostriatum/metabolism , Phenotype , Putamen/metabolism , Rotarod Performance Test , Tyrosine 3-Monooxygenase/metabolism
3.
PLoS One ; 8(4): e60555, 2013.
Article in English | MEDLINE | ID: mdl-23585839

ABSTRACT

Over-expression of phosphoprotein enriched in diabetes/phosphoprotein enriched in astrocytes (PED/PEA-15) causes insulin resistance by interacting with the D4 domain of phospholipase D1 (PLD1). Indeed, the disruption of this association restores insulin sensitivity in cultured cells over-expressing PED/PEA-15. Whether the displacement of PLD1 from PED/PEA-15 improves insulin sensitivity in vivo has not been explored yet. In this work we show that treatment with a recombinant adenoviral vector containing the human D4 cDNA (Ad-D4) restores normal glucose homeostasis in transgenic mice overexpressing PED/PEA-15 (Tg ped/pea-15) by improving both insulin sensitivity and secretion. In skeletal muscle of these mice, D4 over-expression inhibited PED/PEA-15-PLD1 interaction, decreased Protein Kinase C alpha activation and restored insulin induced Protein Kinase C zeta activation, leading to amelioration of insulin-dependent glucose uptake. Interestingly, Ad-D4 administration improved insulin sensitivity also in high-fat diet treated obese C57Bl/6 mice. We conclude that PED/PEA-15-PLD1 interaction may represent a novel target for interventions aiming at improving glucose tolerance.


Subject(s)
Genetic Therapy , Insulin Resistance/genetics , Insulin/metabolism , Obesity/metabolism , Phospholipase D/genetics , Phosphoproteins/genetics , Adenoviridae/genetics , Animals , Apoptosis Regulatory Proteins , Diet, High-Fat/adverse effects , Gene Expression Regulation , Genetic Vectors , Glucose/metabolism , Humans , Insulin Secretion , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/etiology , Obesity/genetics , Obesity/therapy , Phospholipase D/metabolism , Phosphoproteins/metabolism , Protein Binding , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Protein Structure, Tertiary , Signal Transduction , Transgenes
4.
J Appl Toxicol ; 33(6): 451-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22120598

ABSTRACT

Resin-based dental restorative materials release residual monomers that may affect the vitality of pulp cells. The purpose of this study was to evaluate the cytotoxic effect of two light-cured restorative materials with and without bis-GMA resin, respectively (Clearfil Majesty Posterior and Clearfil Majesty Flow) and a self-curing one (Clearfil DC Core Automix) when applied to the fibroblast cell line NIH-3T3. Samples of the materials were light-cured and placed directly in contact to cells for 24, 48, 72 and 96 h. Cytotoxicity was evaluated by measuring cell death by flow cytometry, cell proliferation by proliferation curves analysis and morphological changes by optical microscopy analysis. All the composite materials tested caused a decrease in cell proliferation, albeit at different degrees. However, only Clearfil DC Core Automix induced cell death, very likely by increasing apoptosis. Morphological alteration of treated cells was also evident, particularly in the Clearfil DC Core Automix-treated cells. The different cytotoxic effects of dental composites should be considered when selecting an appropriate resin-based dental restorative material for operative restorations.


Subject(s)
Composite Resins/toxicity , Resins, Synthetic/toxicity , 3T3 Cells , Animals , Annexin A5/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Coloring Agents , Dental Atraumatic Restorative Treatment , Fibroblasts/drug effects , Fibroblasts/ultrastructure , Flow Cytometry , Kinetics , Mice , Necrosis/pathology , Propidium
5.
J Cell Physiol ; 227(4): 1485-92, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21618539

ABSTRACT

Clinical and experimental evidence indicates that atypical antipsychotics impair glucose metabolism. We investigated whether clozapine may directly affect insulin action by analyzing insulin signaling in vitro and in vivo. Clozapine reduced insulin-stimulated glucose uptake in PC12 and in L6 cells, representative models of neuron and skeletal muscle, respectively. Consistently, clozapine reduced insulin effect on insulin receptor (IR) by 40% and on IR substrate-1 (IRS1) tyrosine phosphorylation by 60%. Insulin-stimulated Akt phosphorylation was also reduced by about 40%. Moreover, insulin-dependent phosphorylation of protein kinase C-ζ (PKC-ζ) was completely blunted in clozapine-treated cells. Interestingly, clozapine treatment was accompanied by an insulin-independent increase of Akt phosphorylation, with no change of IR, IRS1, and PKC-ζ basal phosphorylation. The cellular abundance of Ped/Pea-15, an Akt substrate and inducer of insulin resistance, was also increased following clozapine exposure, both in the absence and in the presence of cyclohexymide, a protein synthesis inhibitor. Similar as in cellular models, in the caudate-putamen and in the tibialis muscle of clozapine-treated C57/BL/KsJ mice, Akt phosphorylation and Ped/Pea-15 protein levels were increased and PKC-ζ phosphorylation was decreased. Thus, in these experimental models, clozapine deranged Akt function and up-regulated Ped/Pea-15, thereby inhibiting insulin stimulation of PKC-ζ and of glucose uptake.


Subject(s)
Antipsychotic Agents/pharmacology , Clozapine/pharmacology , Insulin/metabolism , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis Regulatory Proteins , Cell Line , Glucose/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Neurons/drug effects , Neurons/metabolism , PC12 Cells , Phosphorylation , Protein Kinase C/metabolism , Rats , Receptor, Insulin/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects
6.
J Cell Physiol ; 227(5): 2106-16, 2012 May.
Article in English | MEDLINE | ID: mdl-21780113

ABSTRACT

Cell migration is dependent on the control of signaling events that play significant roles in creating contractile force and in contributing to wound closure. We evaluated wound closure in fibroblasts from mice overexpressing (TgPED) or lacking ped/pea-15 (KO), a gene overexpressed in patients with type 2 diabetes. Cultured skin fibroblasts isolated from TgPED mice showed a significant reduction in the ability to recolonize wounded area during scratch assay, compared to control fibroblasts. This difference was observed both in the absence and in the presence of mytomicin C, an inhibitor of mitosis. In time-lapse experiments, TgPED fibroblasts displayed about twofold lower velocity and diffusion coefficient, as compared to controls. These changes were accompanied by reduced spreading and decreased formation of stress fibers and focal adhesion plaques. At the molecular level, TgPED fibroblasts displayed decreased RhoA activation and increased abundance of phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2). Inhibition of ERK1/2 activity by PD98059 restored RhoA activation, cytoskeleton organization and cell motility, and almost completely rescued wound closure of TgPED fibroblasts. Interestingly, skin fibroblasts isolated from KO mice displayed an increased wound closure ability. In vivo, healing of dorsal wounds was delayed in TgPED and accelerated in KO mice. Thus, PED/PEA-15 may affect fibroblast motility by a mechanism, at least in part, mediated by ERK1/2.


Subject(s)
Cell Movement/physiology , Fibroblasts/physiology , Histocompatibility Antigens Class I/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphoproteins/metabolism , Animals , Apoptosis Regulatory Proteins , Cell Adhesion , Cells, Cultured , Diabetes Mellitus, Type 2/physiopathology , Fibroblasts/cytology , Flavonoids/metabolism , Histocompatibility Antigens Class I/genetics , Humans , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Phosphoproteins/genetics , rhoA GTP-Binding Protein/metabolism
7.
Diabetes ; 60(1): 138-47, 2011 01.
Article in English | MEDLINE | ID: mdl-20864515

ABSTRACT

OBJECTIVE: We investigated the function of the Prep1 gene in insulin-dependent glucose homeostasis in liver. RESEARCH DESIGN AND METHODS: Prep1 action on insulin glucoregulatory function has been analyzed in liver of Prep1-hypomorphic mice (Prep1(i/i)), which express 2-3% of Prep1 mRNA. RESULTS: Based on euglycemic hyperinsulinemic clamp studies and measurement of glycogen content, livers from Prep1(i/i) mice feature increased sensitivity to insulin. Tyrosine phosphorylation of both insulin receptor (IR) and insulin receptor substrate (IRS)1/2 was significantly enhanced in Prep1(i/i) livers accompanied by a specific downregulation of the SYP and SHP1 tyrosine phosphatases. Prep1 overexpression in HepG2 liver cells upregulated SYP and SHP1 and inhibited insulin-induced IR and IRS1/2 phosphorylation and was accompanied by reduced glycogen content. Consistently, overexpression of the Prep1 partner Pbx1, but not of p160MBP, mimicked Prep1 effects on tyrosine phosphorylations, glycogen content, and on SYP and SHP1 expression. In Prep1 overexpressing cells, antisense silencing of SHP1, but not that of SYP, rescued insulin-dependent IR phosphorylation and glycogen accumulation. Both Prep1 and Pbx1 bind SHP1 promoter at a site located between nucleotides -2,113 and -1,778. This fragment features enhancer activity and induces luciferase function by 7-, 6-, and 30-fold, respectively, in response to Prep1, Pbx1, or both. CONCLUSIONS: SHP1, a known silencer of insulin signal, is a transcriptional target of Prep1. In liver, transcriptional activation of SHP1 gene by Prep1 attenuates insulin signal transduction and reduces glucose storage.


Subject(s)
Homeodomain Proteins/genetics , Insulin/physiology , Liver/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Animals , Cell Line , DNA Primers , Dietary Fats/pharmacology , Glucose/metabolism , Hep G2 Cells/metabolism , Humans , Liver/enzymology , Liver/metabolism , Mice , Mice, Inbred C57BL , Oligonucleotides/chemistry , Plasmids/genetics , Pre-B-Cell Leukemia Transcription Factor 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transcription Factors/genetics , Transcription, Genetic , Triglycerides/metabolism
8.
Mol Biosyst ; 6(10): 2039-48, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20714510

ABSTRACT

The interaction of Phospholipase D1 (PLD1) by its C-terminal domain D4 with PED/PEA15 has been indicated as a target for type 2 diabetes. PED/PEA15 is overexpressed in several tissues of individuals affected by type 2 diabetes and its overexpression in intact cells and in transgenic animal models impairs insulin regulation of glucose transport by a mechanism mediated by the interaction with D4 and the consequent increase of protein kinase C-alpha activity. Expression of D4 or administration of a peptide mimicking the PED/PEA15 region involved in this interaction to cells stably overexpressing PED/PEA15 reduces its interaction with PLD1, thereby lowering PKC-alpha activation and restoring normal glucose transport mediated by PKC-zeta. By using D4 deletion mutants, we have restricted the PLD1 region involved in PED/PEA15 interaction to an N-terminal fragment named D4alpha (residues 712-818). This region binds PED/PEA15 with the same efficacy as D4 (K(D) approximately 0.7 microM) and, when transfected in different PED/PEA15-overexpressing cells, it is able to reduce PKC-alpha activity and to restore the sensitivity of PKC-zeta to insulin stimulation, independently of the PI3K/Akt signalling. We also show that the effective disruption of the PED/PEA15-PLD1 interaction can restore the normal ERK1/2 signalling. Finally, using a set of overlapping peptides that cover the D4alpha region, we have further restricted the shortest PED/PEA15-binding site to a segment encompassing residues 762-801, suggesting that a quite limited binding interface mostly contributes to the interaction and can thus be a selective target for the design of effective antagonists.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Phospholipase D/metabolism , Phosphoproteins/metabolism , Apoptosis Regulatory Proteins , Base Sequence , DNA Primers , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Mutagenesis, Site-Directed , Phospholipase D/chemistry , Phosphoproteins/chemistry , Protein Binding , Signal Transduction
9.
Hum Gene Ther ; 21(9): 1067-76, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20406097

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive human brain tumor, and is highly resistant to chemo- and radiotherapy. Selectively replicating oncolytic viruses represent a novel approach for the treatment of neoplastic diseases. Coxsackievirus-adenovirus receptor (CAR) is the primary receptor for adenoviruses, and loss or reduction of CAR greatly decreases adenoviral entry. Understanding the mechanisms regulating CAR expression and localization will contribute to increase the efficacy of oncolytic adenoviruses. Two glioma cell lines (U343MG and U373MG) were infected with the oncolytic adenovirus dl922-947. U373MG cells were more susceptible to cell death after viral infection, compared with U343MG cells. The enhanced sensitivity was paralleled by increased adenoviral entry and CAR mRNA and protein levels in U373MG cells. In addition, U373MG cells displayed a decreased ERK1/2 (extracellular signal-regulated kinase-1/2) nuclear-to-cytosolic ratio, compared with U343MG cells. Intracellular content of PED/PEA-15, an ERK1/2-interacting protein, was also augmented in these cells. Both ERK2 overexpression and genetic silencing of PED/PEA-15 by antisense oligonucleotides increased ERK nuclear accumulation and reduced CAR expression and adenoviral entry. Our data indicate that dl922-947 could represent an useful tool for the treatment of GBM and that PED/PEA-15 modulates CAR expression and adenoviral entry, by sequestering ERK1/2.


Subject(s)
Adenoviridae Infections/metabolism , Adenoviridae/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Glioma/enzymology , Glioma/virology , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Receptors, Virus/metabolism , Apoptosis Regulatory Proteins , Cell Death , Cell Line, Tumor , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Down-Regulation , Glioma/pathology , Humans , MAP Kinase Signaling System , Protein Transport , Transfection
10.
Endocrinology ; 150(8): 3584-93, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19359389

ABSTRACT

The AMP-activated protein kinase (AMPK) lies upstream of Akt in the pathway leading to endothelial NO synthase (eNOS) activation. Whether leptin promotes eNOS activation via AMPK-dependent activation of Akt, and which of the two AMPKalpha catalytic subunits is involved, remains unknown. Leptin resistance may be partly attributed to interaction between leptin and C-reactive protein (CRP). We hypothesized that leptin effect on eNOS activation in human aortic endothelial cells might be blunted by direct interaction with human recombinant CRP. Small interfering RNAs (siRNAs) were used to knock down expression of alpha1- or alpha2-AMPK in transient transfection assay to evaluate which is involved in this pathway and whether leptin effect on eNOS activation in human aortic endothelial cells might be blunted by direct interaction with human CRP. siRNA-mediated down-regulation of AMPKalpha1, but not AMPKalpha2, abolished leptin-induced Akt-Ser(473) phosphorylation, eNOS-Ser(1177) phosphorylation, eNOS activation, and cGMP accumulation. By contrast, siRNA-mediated knockdown of Akt1 did not affect AMPKalpha1 phosphorylation, but it abolished leptin-induced phosphorylation of Akt-Ser(473) and eNOS-Ser(1177), suggesting that Akt functions downstream of AMPKalpha1. Preincubation of leptin with human recombinant CRP impaired leptin-induced AMPK activation, eNOS-Ser(1177) phosphorylation, eNOS activity, and intracellular cGMP accumulation. The data are consistent with a model implicating an AMPKalpha1-->Akt-->eNOS pathway leading to NO production in response to leptin supporting the idea that interaction between leptin and CRP may have a role in impairing leptin effect on eNOS activation, suggesting a link between leptin resistance, low-grade inflammation, and endothelial dysfunction.


Subject(s)
AMP-Activated Protein Kinases/metabolism , C-Reactive Protein/metabolism , Leptin/metabolism , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/physiology , Animals , Cell Line , Cyclic GMP/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Female , Humans , Leptin/pharmacology , Mice , Phosphorylation/drug effects , Protein Binding , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/physiology , RNA, Small Interfering , Signal Transduction/drug effects
11.
J Biol Chem ; 283(52): 36088-99, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-18955497

ABSTRACT

Chronic hyperglycemia promotes insulin resistance at least in part by increasing the formation of advanced glycation end products (AGEs). We have previously shown that in L6 myotubes human glycated albumin (HGA) induces insulin resistance by activating protein kinase Calpha (PKCalpha). Here we show that HGA-induced PKCalpha activation is mediated by Src. Coprecipitation experiments showed that Src interacts with both the receptor for AGE (RAGE) and PKCalpha in HGA-treated L6 cells. A direct interaction of PKCalpha with Src and insulin receptor substrate-1 (IRS-1) has also been detected. In addition, silencing of IRS-1 expression abolished HGA-induced RAGE-PKCalpha co-precipitation. AGEs were able to induce insulin resistance also in vivo, as insulin tolerance tests revealed a significant impairment of insulin sensitivity in C57/BL6 mice fed a high AGEs diet (HAD). In tibialis muscle of HAD-fed mice, insulin-induced glucose uptake and protein kinase B phosphorylation were reduced. This was paralleled by a 2.5-fold increase in PKCalpha activity. Similarly to in vitro observations, Src phosphorylation was increased in tibialis muscle of HAD-fed mice, and co-precipitation experiments showed that Src interacts with both RAGE and PKCalpha. These results indicate that AGEs impairment of insulin action in the muscle might be mediated by the formation of a multimolecular complex including RAGE/IRS-1/Src and PKCalpha.


Subject(s)
Glycation End Products, Advanced/metabolism , Insulin/metabolism , Animals , Female , Glucose Tolerance Test , Humans , Hyperglycemia/pathology , Insulin Receptor Substrate Proteins/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Protein Kinase C-alpha/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , src-Family Kinases/metabolism
12.
J Biol Chem ; 283(45): 30970-9, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-18765665

ABSTRACT

Overexpression of the ped/pea-15 gene in mice impairs glucose tolerance and leads to diabetes in conjunction with high fat diet treatment. PED/PEA-15 is also overexpressed in type 2 diabetics as well as in euglycemic offspring from these subjects. The cause(s) of this abnormality remains unclear. In the present work we have cloned and localized the promoter region of the human PED/PEA-15 gene within the first 230 bp of the 5(R)-flanking region. A cis-acting regulatory element located between -320 and -335 bps upstream the PED/PEA-15 gene transcriptional start site (+1) is recognized by both the hepatocyte nuclear factor 4alpha (HNF-4alpha) and the chicken ovalbumin upstream promoter transcription factor II (COUP-TFII), two members of the steroid/thyroid superfamily of transcription factors, both of which are involved in the control of lipid and glucose homeostasis. HNF-4alpha represses PED/PEA-15 expression in HeLa cells, whereas COUP-TFII activates its expression. In hepatocytes, the activation of PED/PEA-15 gene transcription is paralleled by the establishment of a partially dedifferentiated phenotype accompanied by a reduction in mRNA levels encoded by genes normally expressed during liver development. Cotransfection of HeLa cells with a reporter construct containing the PED/PEA-15 response element and various combinations of HNF-4alpha and COUP-TFII expression vectors indicated that COUP-TFII antagonizes the repression of the PED/PEA-15 gene by HNF-4alpha. Thus, at least in part, transcription of the PED/PEA-15 gene in vivo is dependent upon the intracellular balance of these positive and negative regulatory factors. Abnormalities in HNF-4alpha and COUP-TFII balance might have important consequences on glucose tolerance in humans.


Subject(s)
COUP Transcription Factor II/metabolism , Glucose/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Lipid Metabolism/physiology , Phosphoproteins/biosynthesis , Response Elements/physiology , Transcription, Genetic/physiology , Animals , Apoptosis Regulatory Proteins , COUP Transcription Factor II/genetics , Cloning, Molecular , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Glucose/genetics , HeLa Cells , Hepatocyte Nuclear Factor 4/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Liver/metabolism , Mice , Mice, Transgenic , Phosphoproteins/genetics
13.
Mol Cell Biol ; 28(18): 5634-45, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18644868

ABSTRACT

We have examined glucose homeostasis in mice hypomorphic for the homeotic transcription factor gene Prep1. Prep1-hypomorphic (Prep1(i/i)) mice exhibit an absolute reduction in circulating insulin levels but normal glucose tolerance. In addition, these mice exhibit protection from streptozotocin-induced diabetes and enhanced insulin sensitivity with improved glucose uptake and insulin-dependent glucose disposal by skeletal muscle. This muscle phenotype does not depend on reduced expression of the known Prep1 transcription partner, Pbx1. Instead, in Prep1(i/i) muscle, we find normal Pbx1 but reduced levels of the recently identified novel Prep1 interactor p160. Consistent with this reduction, we find a muscle-selective increase in mRNA and protein levels of PGC-1alpha, accompanied by enhanced expression of the GLUT4 transporter, responsible for insulin-stimulated glucose uptake in muscle. Indeed, using L6 skeletal muscle cells, we induced the opposite effects by overexpressing Prep1 or p160, but not Pbx1. In vivo skeletal muscle delivery of p160 cDNA in Prep1(i/i) mice also reverses the molecular phenotype. Finally, we show that Prep1 controls the stability of the p160 protein. We conclude that Prep1 controls insulin sensitivity through the p160-GLUT4 pathway.


Subject(s)
Blood Glucose/metabolism , Carrier Proteins/metabolism , Diabetes Mellitus, Experimental , Homeodomain Proteins/metabolism , Insulin/metabolism , Nuclear Proteins/metabolism , Animals , Carrier Proteins/genetics , DNA-Binding Proteins , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/prevention & control , Female , Glucagon/metabolism , Glucose Tolerance Test , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Homeodomain Proteins/genetics , Homeostasis , Humans , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/metabolism , Nuclear Proteins/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phenotype , Pre-B-Cell Leukemia Transcription Factor 1 , RNA-Binding Proteins , Signal Transduction/physiology , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
14.
J Biol Chem ; 283(31): 21769-78, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18541525

ABSTRACT

Phosphoprotein enriched in diabetes/phosphoprotein enriched in astrocytes (PED/PEA-15) is overexpressed in several tissues of individuals affected by type 2 diabetes. In intact cells and in transgenic animal models, PED/PEA-15 overexpression impairs insulin regulation of glucose transport, and this is mediated by its interaction with the C-terminal D4 domain of phospholipase D1 (PLD1) and the consequent increase of protein kinase C-alpha activity. Here we show that interfering with the interaction of PED/PEA-15 with PLD1 in L6 skeletal muscle cells overexpressing PED/PEA-15 (L6(PED/PEA-15)) restores insulin sensitivity. Surface plasmon resonance and ELISA-like assays show that PED/PEA-15 binds in vitro the D4 domain with high affinity (K(D) = 0.37 +/- 0.13 mum), and a PED/PEA-15 peptide, spanning residues 1-24, PED-(1-24), is able to compete with the PED/PEA-15-D4 recognition. When loaded into L6(PED/PEA-15) cells and in myocytes derived from PED/PEA-15-overexpressing transgenic mice, PED-(1-24) abrogates the PED/PEA-15-PLD1 interaction and reduces protein kinase C-alpha activity to levels similar to controls. Importantly, the peptide restores insulin-stimulated glucose uptake by approximately 70%. Similar results are obtained by expression of D4 in L6(PED/PEA-15). All these findings suggest that disruption of the PED/PEA-15-PLD1 molecular interaction enhances insulin sensitivity in skeletal muscle cells and indicate that PED/PEA-15 as an important target for type 2 diabetes.


Subject(s)
Astrocytes/metabolism , Muscle, Skeletal/metabolism , Phospholipase D/metabolism , Phosphoproteins/metabolism , Animals , Apoptosis Regulatory Proteins , Biological Transport , Gene Deletion , Genetic Vectors , Glucose/metabolism , Mice , Mice, Transgenic , Models, Biological , Muscle, Skeletal/cytology , Peptides/chemistry , Phosphoproteins/genetics , Protein Kinase C-alpha/metabolism , Rats
15.
Diabetes ; 56(3): 622-33, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17327429

ABSTRACT

The phosphoprotein enriched in diabetes/phosphoprotein enriched in astrocytes (ped/pea-15) gene is overexpressed in human diabetes and causes this abnormality in mice. Transgenic mice with beta-cell-specific overexpression of ped/pea-15 (beta-tg) exhibited decreased glucose tolerance but were not insulin resistant. However, they showed impaired insulin response to hyperglycemia. Islets from the beta-tg also exhibited little response to glucose. mRNAs encoding the Sur1 and Kir6.2 potassium channel subunits and their upstream regulator Foxa2 were specifically reduced in these islets. Overexpression of PED/PEA-15 inhibited the induction of the atypical protein kinase C (PKC)-zeta by glucose in mouse islets and in beta-cells of the MIN-6 and INS-1 lines. Rescue of PKC-zeta activity elicited recovery of the expression of the Sur1, Kir6.2, and Foxa2 genes and of glucose-induced insulin secretion in PED/PEA-15-overexpressing beta-cells. Islets from ped/pea-15-null mice exhibited a twofold increased activation of PKC-zeta by glucose; increased abundance of the Sur1, Kir6.2, and Foxa2 mRNAs; and enhanced glucose effect on insulin secretion. In conclusion, PED/PEA-15 is an endogenous regulator of glucose-induced insulin secretion, which restrains potassium channel expression in pancreatic beta-cells. Overexpression of PED/PEA-15 dysregulates beta-cell function and is sufficient to impair glucose tolerance in mice.


Subject(s)
Gene Expression Regulation/physiology , Glucose/pharmacology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Phosphoproteins/metabolism , Potassium Channels/metabolism , Animals , Apoptosis Regulatory Proteins , Cell Line , Down-Regulation , Female , Gene Expression Regulation/drug effects , Insulin Secretion , Male , Mice , Mice, Transgenic , Protein Kinase C/genetics , Protein Kinase C/metabolism
16.
Mol Cell Biol ; 24(11): 5005-15, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15143191

ABSTRACT

Overexpression of the ped/pea-15 gene is a common feature of type 2 diabetes. In the present work, we show that transgenic mice ubiquitously overexpressing ped/pea-15 exhibited mildly elevated random-fed blood glucose levels and decreased glucose tolerance. Treatment with a 60% fat diet led ped/pea-15 transgenic mice to develop diabetes. Consistent with insulin resistance in these mice, insulin administration reduced glucose levels by only 35% after 45 min, compared to 70% in control mice. In vivo, insulin-stimulated glucose uptake was decreased by almost 50% in fat and muscle tissues of the ped/pea-15 transgenic mice, accompanied by protein kinase Calpha activation and block of insulin induction of protein kinase Czeta. These changes persisted in isolated adipocytes from the transgenic mice and were rescued by the protein kinase C inhibitor bisindolylmaleimide. In addition to insulin resistance, ped/pea-15 transgenic mice showed a 70% reduction in insulin response to glucose loading. Stable overexpression of ped/pea-15 in the glucose-responsive MIN6 beta-cell line also caused protein kinase Calpha activation and a marked decline in glucose-stimulated insulin secretion. Antisense block of endogenous ped/pea-15 increased glucose sensitivity by 2.5-fold in these cells. Thus, in vivo, overexpression of ped/pea-15 may lead to diabetes by impairing insulin secretion in addition to insulin action.


Subject(s)
Diabetes Mellitus/genetics , Glucose/metabolism , Histocompatibility Antigens Class I/genetics , Insulin/metabolism , Phosphoproteins/genetics , Animals , Apoptosis Regulatory Proteins , Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Histocompatibility Antigens Class I/biosynthesis , Insulin Secretion , Mice , Mice, Transgenic , Phosphoproteins/biosynthesis
17.
Mol Cell Biol ; 23(13): 4511-21, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12808093

ABSTRACT

The antiapoptotic protein PED/PEA-15 features an Akt phosphorylation motif upstream from Ser(116). In vitro, recombinant PED/PEA-15 was phosphorylated by Akt with a stoichiometry close to 1. Based on Western blotting with specific phospho-Ser(116) PED/PEA-15 antibodies, Akt phosphorylation of PED/PEA-15 occurred mainly at Ser(116). In addition, a mutant of PED/PEA-15 featuring the substitution of Ser(116)-->Gly (PED(S116-->G)) showed 10-fold-decreased phosphorylation by Akt. In intact 293 cells, Akt also induced phosphorylation of PED/PEA-15 at Ser(116). Based on pull-down and coprecipitation assays, PED/PEA-15 specifically bound Akt, independently of Akt activity. Serum activation of Akt as well as BAD phosphorylation by Akt showed no difference in 293 cells transfected with PED/PEA-15 and in untransfected cells (which express no endogenous PED/PEA-15). However, the antiapoptotic action of PED/PEA-15 was almost twofold reduced in PED(S116-->G) compared to that in PED/PEA-15(WT) cells. PED/PEA-15 stability closely paralleled Akt activation by serum in 293 cells. In these cells, the nonphosphorylatable PED(S116-->G) mutant exhibited a degradation rate threefold greater than that observed with wild-type PED/PEA-15. In the U373MG glioma cells, blocking Akt also reduced PED/PEA-15 levels and induced sensitivity to tumor necrosis factor-related apoptosis-inducing ligand apoptosis. Thus, phosphorylation by Akt regulates the antiapoptotic function of PED/PEA-15 at least in part by controlling the stability of PED/PEA-15. In part, Akt survival signaling may be mediated by PED/PEA-15.


Subject(s)
Apoptosis , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Androstadienes/pharmacology , Apoptosis Regulatory Proteins , Binding Sites , Blotting, Western , Cell Line , Cycloheximide/pharmacology , DNA, Complementary/metabolism , Enzyme Inhibitors/pharmacology , Glioma/metabolism , Glutathione Transferase/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Mutation , Peptides/chemistry , Phosphorylation , Plasmids/metabolism , Protein Binding , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt , Recombinant Proteins/metabolism , Serine/metabolism , Signal Transduction , Time Factors , Transfection , Tumor Cells, Cultured , Wortmannin
18.
J Biol Chem ; 277(13): 11013-8, 2002 Mar 29.
Article in English | MEDLINE | ID: mdl-11790785

ABSTRACT

293 kidney embryonic cells feature very low levels of the anti-apoptotic protein PED. In these cells, expression of PED to levels comparable with those occurring in normal adult cells inhibits apoptosis induced by growth factor deprivation and by exposure to H(2)O(2) or anisomycin. In PED-expressing 293 cells (293(PED)), inhibition of apoptosis upon growth factor deprivation was paralleled by decreased phosphorylation of JNK1/2. In 293(PED) cells, decreased apoptosis induced by anisomycin and H(2)O(2) was also accompanied by block of JNK1/2 and p38 phosphorylations, respectively. Impaired activity of these stress kinases by PED correlated with inhibition of stress-induced Cdc-42, MKK4, and MKK6 activation. At variance with JNK1/2 and p38, PED expression increased basal and growth factor-stimulated Ras-Raf-1 co-precipitation and MAPK phosphorylation and activity. Treatment of 293(PED) cells with the MEK inhibitor PD98059 blocked ERK1/2 phosphorylations with no effect on inhibition of JNK1/2 and p38 activities. Complete rescue of JNK and p38 functions in 293(PED) cells by overexpressing JNK1 or p38, respectively, enabled only partial recovery of apoptotic response to growth factor deprivation and anisomycin. However, simultaneous rescue of JNK and p38 activities accompanied by block of ERK1/2 fully restored these responses. Thus, PED controls activity of the ERK, JNK, and p38 subfamilies of MAPKs. PED anti-apoptotic function in the 293 cells requires PED simultaneous activation of ERK1/2 and inhibition of the JNK/p38 signaling systems by PED.


Subject(s)
Apoptosis/physiology , Mitogen-Activated Protein Kinases/metabolism , Phosphoproteins/physiology , Animals , Apoptosis Regulatory Proteins , Base Sequence , CHO Cells , Cell Line , Cricetinae , DNA Primers , Humans , Intracellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Phosphorylation , Signal Transduction , cdc42 GTP-Binding Protein/metabolism , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL