Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
1.
Oncogene ; 34(25): 3296-304, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25151967

ABSTRACT

Isoprenylcysteine carboxylmethyltransferase (Icmt) catalyzes the last of the three-step posttranslational protein prenylation process for the so-called CaaX proteins, which includes many signaling proteins, such as most small GTPases. Despite extensive studies on Icmt and its regulation of cell functions, the mechanisms of much of the impact of Icmt on cellular functions remain unclear. Our recent studies demonstrated that suppression of Icmt results in induction of autophagy, inhibition of cell growth and inhibition of proliferation in various cancer cell types, prompting this investigation of potential metabolic regulation by Icmt. We report here the findings that Icmt inhibition reduces the function of mitochondrial oxidative phosphorylation in multiple cancer cell lines. In-depth oximetry analysis demonstrated that functions of mitochondrial complex I, II and III are subject to Icmt regulation. Consistently, Icmt inhibition decreased cellular ATP and depleted critical tricarboxylic acid cycle metabolites, leading to suppression of cell anabolism and growth, and marked autophagy. Several different approaches demonstrated that the impact of Icmt inhibition on cell proliferation and viability was largely mediated by its effect on mitochondrial respiration. This previously unappreciated function of Icmt, which can be therapeutically exploited, likely has a significant role in the impact of Icmt on tumorigenic processes.


Subject(s)
Mitochondria/metabolism , Protein Methyltransferases/metabolism , AMP-Activated Protein Kinases/metabolism , Carcinogenesis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Respiration/drug effects , Cell Survival/drug effects , Electron Transport Chain Complex Proteins/metabolism , Energy Metabolism/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Indoles/pharmacology , Mitochondria/drug effects , Mitochondria/enzymology , Protein Methyltransferases/antagonists & inhibitors
2.
Mitochondrion ; 20: 95-102, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25530493

ABSTRACT

Single-gene mutations that disrupt mitochondrial respiratory chain function in Caenorhabditis elegans change patterns of protein expression and metabolites. Our goal was to develop useful molecular fingerprints employing adaptable techniques to recognize mitochondrial defects in the electron transport chain. We analyzed mutations affecting complex I, complex II, or ubiquinone synthesis and discovered overarching patterns in the response of C. elegans to mitochondrial dysfunction across all of the mutations studied. These patterns are in KEGG pathways conserved from C. elegans to mammals, verifying that the nematode can serve as a model for mammalian disease. In addition, specific differences exist between mutants that may be useful in diagnosing specific mitochondrial diseases in patients.


Subject(s)
Caenorhabditis elegans/chemistry , Electron Transport Chain Complex Proteins/genetics , Metabolome , Mitochondria/enzymology , Mutation , Proteome/analysis , Animals , Caenorhabditis elegans Proteins/analysis , Caenorhabditis elegans Proteins/genetics , Mitochondrial Proteins/genetics
3.
Diabetologia ; 55(2): 321-30, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22065088

ABSTRACT

AIMS/HYPOTHESIS: Insulin resistance (IR) improves with weight loss, but this response is heterogeneous. We hypothesised that metabolomic profiling would identify biomarkers predicting changes in IR with weight loss. METHODS: Targeted mass spectrometry-based profiling of 60 metabolites, plus biochemical assays of NEFA, ß-hydroxybutyrate, ketones, insulin and glucose were performed in baseline and 6 month plasma samples from 500 participants who had lost ≥4 kg during Phase I of the Weight Loss Maintenance (WLM) trial. Homeostatic model assessment of insulin resistance (HOMA-IR) and change in HOMA-IR with weight loss (∆HOMA-IR) were calculated. Principal components analysis (PCA) and mixed models adjusted for race, sex, baseline weight, and amount of weight loss were used; findings were validated in an independent cohort of patients (n = 22). RESULTS: Mean weight loss was 8.67 ± 4.28 kg; mean ∆HOMA-IR was -0.80 ± 1.73, range -28.9 to 4.82). Baseline PCA-derived factor 3 (branched chain amino acids [BCAAs] and associated catabolites) correlated with baseline HOMA-IR (r = 0.50, p < 0.0001) and independently associated with ∆HOMA-IR (p < 0.0001). ∆HOMA-IR increased in a linear fashion with increasing baseline factor 3 quartiles. Amount of weight loss was only modestly correlated with ∆HOMA-IR (r = 0.24). These findings were validated in the independent cohort, with a factor composed of BCAAs and related metabolites predicting ∆HOMA-IR (p = 0.007). CONCLUSIONS/INTERPRETATION: A cluster of metabolites comprising BCAAs and related analytes predicts improvement in HOMA-IR independent of the amount of weight lost. These results may help identify individuals most likely to benefit from moderate weight loss and elucidate novel mechanisms of IR in obesity.


Subject(s)
Amino Acids, Branched-Chain/chemistry , Insulin Resistance , Adult , Algorithms , Amino Acids/chemistry , Biomarkers/metabolism , Body Mass Index , Body Weight , Cohort Studies , Female , Humans , Male , Mass Spectrometry/methods , Middle Aged , Principal Component Analysis , Weight Loss
4.
J Neuroendocrinol ; 22(3): 175-80, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20059648

ABSTRACT

In our previous study, moderate increases in plasma leptin levels achieved via administration of recombinant adenovirus containing the rat leptin cDNA were shown to correct the abnormal metabolic profile in rats with diet-induced obesity, suggesting that these animals had developed resistance to the metabolic effects of leptin, which could be reversed by leptin gene over-expression. However, the effect of this therapeutic strategy on blood pressure was not investigated. The present study aimed to determine whether a moderate increase of endogenous plasma leptin levels affected arterial blood pressure in rats with diet-induced obesity and hypertension. The major finding from the present study was that the natural rise in plasma leptin with weight-gain is insufficient to counterbalance high blood pressure associated with obesity, additional increases of circulating leptin levels with adenoviral leptin gene therapy led to normalisation of blood pressure in high-fat diet-induced obese and hypertensive rats. Mechanistically, the reduction of blood pressure by leptin in obese rats was likely independent of alpha-adrenergic and acetylcholinergic receptor mediation. This is the first study to demonstrate that further increases in circulating leptin levels by leptin gene transfer during obesity could reduce blood pressure.


Subject(s)
Genetic Therapy , Hypertension/therapy , Leptin/biosynthesis , Obesity/complications , Adenoviridae , Animals , Dietary Fats/administration & dosage , Gene Transfer Techniques , Hypertension/etiology , Leptin/blood , Leptin/genetics , Male , Obesity/genetics , Obesity/metabolism , Rats , Rats, Wistar
5.
Diabetologia ; 53(4): 757-67, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20076942

ABSTRACT

AIMS/HYPOTHESIS: Insulin resistance (IR) is associated with obesity, but can also develop in individuals with normal body weight. We employed comprehensive profiling methods to identify metabolic events associated with IR, while controlling for obesity. METHODS: We selected 263 non-obese (BMI approximately 24 kg/m2) Asian-Indian and Chinese men from a large cross-sectional study carried out in Singapore. Individuals taking medication for diabetes or hyperlipidaemia were excluded. Participants were separated into lower and upper tertiles of IR based on HOMA indices of < or =1.06 or > or =1.93, respectively. MS-based metabolic profiling of acylcarnitines, amino acids and organic acids was combined with hormonal and cytokine profiling in all participants. RESULTS: After controlling for BMI, commonly accepted risk factors for IR, including circulating fatty acids and inflammatory cytokines, did not discriminate the upper and lower quartiles of insulin sensitivity in either Asian- Indian or Chinese men. Instead, IR was correlated with increased levels of alanine, proline, valine, leucine/isoleucine, phenylalanine, tyrosine, glutamate/glutamine and ornithine, and a cluster of branched-chain and related amino acids identified by principal components analysis. These changes were not due to increased protein intake by individuals in the upper quartile of IR. Increased abdominal adiposity and leptin, and decreased adiponectin and IGF-binding protein 1 were also correlated with IR. CONCLUSIONS/INTERPRETATION: These findings demonstrate that perturbations in amino acid homeostasis, but not inflammatory markers or NEFAs, are associated with IR in individuals of relatively low body mass.


Subject(s)
Body Mass Index , Insulin Resistance/physiology , Adiponectin/blood , Adult , Amino Acids/blood , Amino Acids/metabolism , Asian People , Blood Glucose/metabolism , Cholesterol/blood , Cross-Sectional Studies , Demography , Granulocyte-Macrophage Colony-Stimulating Factor/blood , Humans , India , Insulin/blood , Insulin-Like Growth Factor Binding Proteins/blood , Interleukins/blood , Life Style , Lipids/blood , Male , Mass Spectrometry , Middle Aged , Patient Selection , Racial Groups , Risk Factors , Singapore/epidemiology , White People
6.
Int J Obes Relat Metab Disord ; 27 Suppl 3: S12-6, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14704737

ABSTRACT

Inflammation plays an important role in the destruction of pancreatic islet beta-cells that leads to type I diabetes. This involves infiltration of T-cells and macrophages into the islets and local production of inflammatory cytokines such as interleukin (IL)-1 beta, tumor necrosis factor (TNF)-alpha, and interferon (IFN)-gamma. Our laboratory has developed several strategies for protecting beta-cells against oxidative stress and cytokine-induced cytotoxicity. These include a cytokine selection strategy that results in cell lines that are resistant to the combined effects of IL-1 beta+IFN-gamma. More recently, we have combined the cytokine selection procedure with overexpression of the antiapoptotic gene bcl-2, resulting in cell lines with greater resistance to oxidative stress and cytokine-induced damage than achieved with either procedure alone. This article summarizes this work and the remarkably divergent mechanisms by which protection is achieved in the different model systems. We also discuss the potential relevance of insights gained from these approaches for enhancing islet cell survival and function in both major forms of diabetes.


Subject(s)
Cytokines/pharmacology , Diabetes Mellitus/pathology , Insulinoma/pathology , Islets of Langerhans/pathology , Pancreatic Neoplasms/pathology , Animals , Apoptosis/genetics , Cell Survival/drug effects , Humans , Islets of Langerhans/drug effects , Tumor Cells, Cultured
7.
Nature ; 413(6852): 131-8, 2001 Sep 13.
Article in English | MEDLINE | ID: mdl-11557972

ABSTRACT

Blood glucose levels are maintained by the balance between glucose uptake by peripheral tissues and glucose secretion by the liver. Gluconeogenesis is strongly stimulated during fasting and is aberrantly activated in diabetes mellitus. Here we show that the transcriptional coactivator PGC-1 is strongly induced in liver in fasting mice and in three mouse models of insulin action deficiency: streptozotocin-induced diabetes, ob/ob genotype and liver insulin-receptor knockout. PGC-1 is induced synergistically in primary liver cultures by cyclic AMP and glucocorticoids. Adenoviral-mediated expression of PGC-1 in hepatocytes in culture or in vivo strongly activates an entire programme of key gluconeogenic enzymes, including phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase, leading to increased glucose output. Full transcriptional activation of the PEPCK promoter requires coactivation of the glucocorticoid receptor and the liver-enriched transcription factor HNF-4alpha (hepatic nuclear factor-4alpha) by PGC-1. These results implicate PGC-1 as a key modulator of hepatic gluconeogenesis and as a central target of the insulin-cAMP axis in liver.


Subject(s)
Blood Glucose/metabolism , DNA-Binding Proteins , Gluconeogenesis , Liver/metabolism , Transcription Factors/physiology , 3T3 Cells , Amino Acid Motifs , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cell Line , Cyclic AMP/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/metabolism , Fasting , Hepatocyte Nuclear Factor 4 , Hormones/metabolism , Insulin/physiology , Male , Mice , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Phosphoproteins/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Receptors, Glucocorticoid/metabolism , Response Elements , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured
8.
Science ; 293(5531): 818-9, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11486079

ABSTRACT

What do the regulation of translation initiation and glucose metabolism have to do with each other? Quite a lot, it seems, according to Sonenberg and Newgard in their Perspective. They discuss new findings that identify the kinase responsible for inactivating eIF2--a factor that is required for translation initiation (and hence protein synthesis)--when the endoplasmic reticulum is under stress. Loss of this kinase results in destruction of insulin-producing b cells in the pancreas and dysregulation of glucose homeostasis.


Subject(s)
Endoplasmic Reticulum/metabolism , Eukaryotic Initiation Factor-2/metabolism , Glucose/metabolism , Protein Biosynthesis , eIF-2 Kinase/metabolism , Animals , Blood Glucose/metabolism , Gluconeogenesis , Homeostasis , Hyperglycemia/etiology , Hypoglycemia/etiology , Islets of Langerhans/enzymology , Islets of Langerhans/metabolism , Liver/metabolism , Mice , Mutation , Phosphorylation , Protein Folding
9.
J Biol Chem ; 276(14): 10722-9, 2001 Apr 06.
Article in English | MEDLINE | ID: mdl-11148207

ABSTRACT

The final step of gluconeogenesis and glycogenolysis is catalyzed by the glucose-6-phosphatase (Glc-6-Pase) enzyme complex, located in the endoplasmic reticulum. The complex consists of a 36-kDa catalytic subunit (P36), a 46-kDa glucose 6-phosphate translocase (P46), and putative glucose and inorganic phosphate transporters. Mutations in the genes encoding P36 or P46 have been linked to glycogen storage diseases type Ia and type Ib, respectively. However, the relative roles of these two proteins in control of the rate of glucose 6-phosphate hydrolysis have not been defined. To gain insight into this area, we have constructed a recombinant adenovirus containing the cDNA encoding human P46 (AdCMV-P46) and treated rat hepatocytes with this virus, or a virus encoding P36 (AdCMV-P36), or the combination of both viruses, resulting in large and equivalent increases in expression of the transgenes within 8-24 h of viral treatment. The overexpressed P46 protein was appropriately targeted to hepatocyte microsomes and caused a 58% increase in glucose 6-phosphate hydrolysis in nondetergent-treated (intact) microsomal preparations relative to controls, whereas overexpression of P36 caused a 3.6-fold increase. Overexpression of P46 caused a 50% inhibition of glycogen accumulation in hepatocytes from fasted rats incubated at 25 mm glucose relative to cells treated with a control virus (AdCMV-betaGAL). Furthermore, in hepatocytes from fed rats cultured at 25 mm glucose and then exposed to 15 mm glucose, AdCMV-P46 treatment activated glycogenolysis, as indicated by a 50% reduction in glycogen content relative to AdCMV-betaGAL-treated controls. In contrast, overexpression of P46 had only small effects on glycolysis, whereas overexpression of P36 had large effects on both glycogen metabolism and glycolysis, even in the presence of co-overexpressed glucokinase. Finally, P46 overexpression enhanced glucose 1-phosphate but not fructose 6-phosphate hydrolysis in intact microsomes, providing a mechanism by which P46 overexpression may exert its preferential effects on glycogen metabolism.


Subject(s)
Gene Expression Regulation, Enzymologic , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Glycogen/metabolism , Liver/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Animals , Biological Transport/genetics , Humans , Male , Rats
10.
J Clin Invest ; 107(1): 91-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11134184

ABSTRACT

Hepatic 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase is an important regulatory enzyme of glucose metabolism. By controlling the level of fructose-2,6-bisphosphate, an allosteric activator of the glycolytic enzyme 6-phosphofructo-1-kinase and an inhibitor of the gluconeogenic enzyme fructose-1,6-bisphosphatase, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase regulates hepatic glucose output. We studied the effects of adenovirus-mediated overexpression of this enzyme on hepatic glucose metabolism in normal or diabetic mice. These animals were treated with virus encoding either wild-type or bisphosphatase activity-deficient 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase. Seven days after virus injection, hepatic fructose-2,6-bisphosphate levels increased significantly in both normal and diabetic mice, with larger increases observed in animals with overexpression of the mutant enzyme. Blood glucose levels in normal mice overexpressing either enzyme were lowered, accompanied by increased plasma lactate, triglycerides, and FFAs. Blood glucose levels were markedly reduced in diabetic mice overexpressing the wild-type enzyme, and still more so in mice overexpressing the mutant form of the enzyme. The lower blood glucose levels in diabetic mice were accompanied by partially normalized plasma triglycerides and FFAs, increased plasma lactate, and increased liver glycogen levels, relative to diabetic mice treated with a control adenovirus. Our findings underscore the critical role played by hepatic 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase in control of fuel homeostasis and suggest that this enzyme may be considered as a therapeutic target in diabetes.


Subject(s)
Blood Glucose/metabolism , Glucose/biosynthesis , Liver/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Adenoviridae/genetics , Animals , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/metabolism , Gene Expression , Genetic Vectors , Liver Glycogen/metabolism , Male , Mice , Mutation , Phosphofructokinase-2 , Phosphoric Monoester Hydrolases/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics
11.
J Biol Chem ; 276(1): 766-72, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11024034

ABSTRACT

Destruction of pancreatic islet beta-cells in type 1 diabetes appears to result from direct contact with infiltrating T-cells and macrophages and exposure to inflammatory cytokines such as interferon (IFN)-gamma, interleukin (IL)-1 beta, and tumor necrosis factor TNF-alpha that such cells produce. We recently reported on a method for selection of insulinoma cells that are resistant to the cytotoxic effects of inflammatory cytokines (INS-1(res)), involving their growth in progressively increasing concentrations of IL-1 beta plus IFN-gamma, and selection of surviving cells. In the current study, we have investigated the molecular mechanism of cytokine resistance in INS-1(res) cells. By focusing on the known components of the IFN-gamma receptor signaling pathway, we have discovered that expression levels of signal transducer and activator of transcription (STAT)-1 alpha are closely correlated with the cytokine-resistant and -sensitive phenotypes. That STAT-1 alpha is directly involved in development of cytokine resistance is demonstrated by an increase of viability from 10 +/- 2% in control cells to 50 +/- 6% in cells with adenovirus-mediated overexpression of STAT-1 alpha (p < 0.001) after culture of both cell groups in the presence of 100 units/ml IFN-gamma plus 10 ng/ml IL-1 beta for 48 h. The resistance to IL-1 beta plus IFN-gamma in STAT-1 alpha-expressing cells is due in part to interference with IL-1 beta-mediated stimulation of inducible nitric-oxide synthase expression and nitric oxide production. Furthermore, overexpression of STAT-1 alpha does not impair robust glucose-stimulated insulin secretion in the INS-1-derived cell line 832/13. We conclude that expression of STAT-1 alpha may be a means of protecting insulin-producing cell lines from cytokine damage, which, in conjunction with appropriate cell-impermeant macroencapsulation devices, may allow such cells to be used for insulin replacement in type 1 diabetes.


Subject(s)
Cytokines/antagonists & inhibitors , Cytokines/toxicity , Drug Resistance, Neoplasm , Insulinoma/metabolism , Transcription Factors/metabolism , Adenoviridae/genetics , Animals , Cell Survival/drug effects , Cytokines/pharmacology , Gene Expression Regulation/drug effects , Glucose/pharmacology , Humans , Insulin/metabolism , Insulin Secretion , Interferon-Stimulated Gene Factor 3 , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Phosphorylation , Rats , Receptors, Interferon/metabolism , Signal Transduction/drug effects , Transcription Factors/genetics , Transfection , Tumor Cells, Cultured
12.
J Biol Chem ; 276(9): 6479-84, 2001 Mar 02.
Article in English | MEDLINE | ID: mdl-11113153

ABSTRACT

The long-chain acyl-CoA (LC-CoA) model of glucose-stimulated insulin secretion (GSIS) holds that secretion is linked to a glucose-induced increase in malonyl-CoA level and accumulation of LC-CoA in the cytosol. We have previously tested the validity of this proposal by overexpressing goose malonyl-CoA decarboxylase (MCD) in INS-1 cells, but these studies have been criticized due to: 1) the small insulin secretion response (2-4-fold) of the INS-1 cells used; 2) unknown contribution of the ATP-sensitive K(+) (K(ATP)) channel-independent pathway of GSIS in INS-1 cells, which has been implicated as the site at which lipids regulate insulin granule exocytosis; and 3) deletion of the N-terminal mitochondrial targeting sequence, but not the C-terminal peroxisomal targeting sequence in the goose MCD construct, raising the possibility that a significant fraction of the overexpressed enzyme was localized to peroxisomes. To address these outstanding concerns, INS-1-derived 832/13 cells, which exhibit robust K(ATP) channel-dependent and -independent pathways of GSIS, were treated with a new adenovirus encoding human MCD lacking both its mitochondrial and peroxisomal targeting sequences (AdCMV-MCD Delta 5), resulting in large increases in cytosolic MCD activity. Treatment of 832/13 cells with AdCMV-MCD Delta 5 completely blocked the glucose-induced rise in malonyl-CoA and attenuated the inhibitory effect of glucose on fatty acid oxidation. However, MCD overexpression had no effect on K(ATP) channel-dependent or -independent GSIS in 832/13 cells. Furthermore, combined treatment of 832/13 cells with AdCMV-MCD Delta 5 and triacsin C, an inhibitor of long chain acyl-CoA synthetase that reduces LC-CoA levels, did not impair GSIS. These findings extend our previous observations and are not consistent with the LC-CoA hypothesis as originally set forth.


Subject(s)
Carboxy-Lyases/physiology , Glucose/pharmacology , Insulin/metabolism , Islets of Langerhans/physiology , Malonyl Coenzyme A/biosynthesis , Adenoviridae/genetics , Cell Line , Diazoxide/pharmacology , Fatty Acids/metabolism , Humans , Insulin Secretion , Oxidation-Reduction , Triazenes/pharmacology
13.
Diabetes ; 49(12): 1967-77, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11117996

ABSTRACT

Glucose is stored in mammalian tissues in the form of glycogen. Glycogen levels are markedly reduced in liver or muscle cells of patients with insulin-resistant or insulin-deficient forms of diabetes, suggesting that impaired glycogen synthesis may contribute to development of hyperglycemia. Recently, interest in this area has been further stimulated by new insights into the spatial organization of metabolic enzymes within cells and the importance of such organization in regulation of glycogen metabolism. It is now clear that a four-member family of glycogen targeting subunits of protein phosphatase-1 (PP1) plays a major role in coordinating these events. These proteins target PP1 to the glycogen particle and also bind differentially to glycogen synthase, glycogen phosphorylase, and phosphorylase kinase, thereby serving as molecular scaffolds. Moreover, the various glycogen-targeting subunits have distinct tissue expression patterns and can influence regulation of glycogen metabolism in response to glycogenic and glycogenolytic signals. The purpose of this article is to summarize new insights into the structure, function, regulation, and metabolic effects of the glycogen-targeting subunits of PP1 and to evaluate the possibility that these molecules could serve as therapeutic targets for lowering of blood glucose in diabetes.


Subject(s)
Glucose/metabolism , Phosphoprotein Phosphatases/metabolism , Animals , Diabetes Mellitus/drug therapy , Glycogen/metabolism , Humans , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/therapeutic use , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/therapeutic use , Protein Phosphatase 1 , Structure-Activity Relationship
14.
J Biol Chem ; 275(52): 40817-26, 2000 Dec 29.
Article in English | MEDLINE | ID: mdl-11007796

ABSTRACT

Glucose serves as both a nutrient and regulator of physiological and pathological processes. Presently, we found that glucose and certain sugars rapidly activated extracellular signal-regulated kinase (ERK) by a mechanism that was: (a) independent of glucose uptake/metabolism and protein kinase C but nevertheless cytochalasin B-inhibitable; (b) dependent upon proline-rich tyrosine kinase-2 (PYK2), GRB2, SOS, RAS, RAF, and MEK1; and (c) amplified by overexpression of the Glut1, but not Glut2, Glut3, or Glut4, glucose transporter. This amplifying effect was independent of glucose uptake but dependent on residues 463-468, IASGFR, in the Glut1 C terminus. Accordingly, glucose effects on ERK were amplified by expression of Glut4/Glut1 or Glut2/Glut1 chimeras containing IASGFR but not by Glut1/Glut4 or Glut1/Glut2 chimeras lacking these residues. Also, deletion of Glut1 residues 469-492 was without effect, but mutations involving serine 465 or arginine 468 yielded dominant-negative forms that inhibited glucose-dependent ERK activation. Glucose stimulated the phosphorylation of tyrosine residues 402 and 881 in PYK2 and binding of PYK2 to Myc-Glut1. Our findings suggest that: (a) glucose activates the GRB2/SOS/RAS/RAF/MEK1/ERK pathway by a mechanism that requires PYK2 and residues 463-468, IASGFR, in the Glut1 C terminus and (b) Glut1 serves as a sensor, transducer, and amplifier for glucose signaling to PYK2 and ERK.


Subject(s)
Glucose/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Monosaccharide Transport Proteins/physiology , Protein-Tyrosine Kinases/physiology , 3T3 Cells , Adipocytes/metabolism , Animals , Deoxyglucose/metabolism , Disaccharides/pharmacology , Focal Adhesion Kinase 2 , Glucose Transporter Type 1 , Glucose Transporter Type 2 , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Rats
15.
J Biol Chem ; 275(46): 35799-806, 2000 Nov 17.
Article in English | MEDLINE | ID: mdl-10967113

ABSTRACT

To better understand the action of glucose on fatty acid metabolism in the beta-cell and the link between chronically elevated glucose or fatty acids and beta-cell decompensation in adipogenic diabetes, we investigated whether glucose regulates peroxisomal proliferator-activated receptor (PPAR) gene expression in the beta-cell. Islets or INS(832/13) beta-cells exposed to high glucose show a 60-80% reduction in PPARalpha mRNA expression. Oleate, either in the absence or presence of glucose, has no effect. The action of glucose is dose-dependent in the 6-20 mm range and maximal after 6 h. Glucose also causes quantitatively similar reductions in PPARalpha protein and DNA binding activity of this transcription factor. The effect of glucose is blocked by the glucokinase inhibitor mannoheptulose, is partially mimicked by 2-deoxyglucose, and is not blocked by the 3-O-methyl or the 6-deoxy analogues of the sugar that are not phosphorylated. Chronic elevated glucose reduces the expression levels of the PPAR target genes, uncoupling protein 2 and acyl-CoA oxidase, which are involved in fat oxidation and lipid detoxification. A 3-day exposure of INS-1 cells to elevated glucose results in a permanent rise in malonyl-CoA, the inhibition of fat oxidation, and the promotion of fatty acid esterification processes and causes elevated insulin secretion at low glucose. The results suggest that a reduction in PPARalpha gene expression together with a rise in malonyl-CoA plays a role in the coordinated adaptation of beta-cell glucose and lipid metabolism to hyperglycemia and may be implicated in the mechanism of beta-cell "glucolipotoxicity."


Subject(s)
Down-Regulation/drug effects , Glucose/pharmacology , Islets of Langerhans/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Glucose/analogs & derivatives , Glucose/metabolism , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Kinetics , Malonyl Coenzyme A/metabolism , Models, Biological , Oleic Acid/metabolism , Oleic Acid/pharmacology , Oxidation-Reduction/drug effects , Palmitic Acid/metabolism , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/metabolism , Response Elements/drug effects , Transcription Factors/metabolism , Triglycerides/metabolism
16.
J Biol Chem ; 275(51): 39991-5, 2000 Dec 22.
Article in English | MEDLINE | ID: mdl-10998419

ABSTRACT

There is growing evidence that glycogen targeting subunits of protein phosphatase-1 play a critical role in regulation of glycogen metabolism. In the current study, we have investigated the effects of adenovirus-mediated overexpression of a specific glycogen targeting subunit known as protein targeting to glycogen (PTG) in cultured human muscle cells. PTG was overexpressed both in muscle cells cultured at high glucose (glycogen replete) or in cells incubated for 18 h in the absence of glucose and then incubated in high glucose (glycogen re-synthesizing). In both glycogen replete and glycogen resynthesizing cells, PTG overexpression caused glycogen to be synthesized at a linear rate 1-5 days after viral treatment, while in cells treated with a virus lacking a cDNA insert (control virus), glycogen content reached a plateau at day 1 with no further increase. In the glycogen replete PTG overexpressing cells, glycogen content was 20 times that in controls at day 5. Furthermore, in cells undergoing glycogen resynthesis, PTG overexpression caused a doubling of the initial rate of glycogen synthesis over the first 24 h relative to cells treated with control virus. In both sets of experiments, the effects of PTG on glycogen synthesis were correlated with a 2-3-fold increase in glycogen synthase activity state, with no changes in glycogen phosphorylase activity. The alterations in glycogen synthase activity were not accompanied by changes in the intracellular concentration of glucose 6-phosphate. We conclude that PTG overexpression activates glycogen synthesis in a glucose 6-phosphate-independent manner in human muscle cells while overriding glycogen-mediated inhibition. Our findings suggest that modulation of PTG expression in muscle may be a mechanism for enhancing muscle glucose disposal and improving glucose tolerance in diabetes.


Subject(s)
Glucose-6-Phosphate/metabolism , Glycogen/metabolism , Muscle Proteins/metabolism , Muscles/metabolism , Cells, Cultured , Glycogen/biosynthesis , Humans , Muscles/cytology , Muscles/enzymology , Phosphorylases/metabolism
17.
J Biol Chem ; 275(34): 26396-403, 2000 Aug 25.
Article in English | MEDLINE | ID: mdl-10862764

ABSTRACT

Glycogen-targeting subunits of protein phosphatase-1 facilitate interaction of the phosphatase with enzymes of glycogen metabolism. We have shown that overexpression of one member of the family, protein targeting to glycogen (PTG), causes large increases in glycogen storage in isolated hepatocytes or intact rat liver. In the current study, we have compared the metabolic and regulatory properties of PTG (expressed in many tissues), with two other members of the gene family, G(L) (expressed primarily in liver) and G(M)/R(Gl) (expressed primarily in striated muscle). Adenovirus-mediated expression of these proteins in hepatocytes led to the following key observations. 1) G(L) has the highest glycogenic potency among the three forms studied. 2) Glycogen synthase activity ratio is much higher in G(L)-overexpressing cells than in PTG or G(M)/R(Gl)-overexpressing cells. Thus, at moderate levels of G(L) overexpression, glycogen synthase activity is increased by insulin treatment, but at higher levels of G(L) expression, insulin is no longer required to achieve maximal synthase activity. In contrast, cells with high levels of PTG overexpression retain dose-dependent regulation of glycogen synthesis and glycogen synthase enzyme activity by insulin. 3) G(L)- and G(M)/R(Gl)-overexpressing cells exhibit a strong glycogenolytic response to forskolin, whereas PTG-overexpressing cells are less responsive. This difference may be explained in part by a lesser forskolin-induced increase in glycogen phosphorylase activity in PTG-overexpressing cells. Based on these results, we suggest that expression of either G(L) or G(M)/R(Gl) in liver of diabetic animals may represent a strategy for lowering of blood glucose levels in diabetes.


Subject(s)
Carrier Proteins/metabolism , Liver/metabolism , Phosphoprotein Phosphatases/metabolism , Animals , Cells, Cultured , Colforsin/pharmacology , Glycogen/metabolism , Glycogen Synthase/metabolism , Intracellular Signaling Peptides and Proteins , Male , Phosphorylases/metabolism , Protein Phosphatase 1 , Rats , Rats, Wistar , Structure-Activity Relationship
18.
Diabetes ; 49(4): 562-70, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10871193

ABSTRACT

Engineered insulinoma cell lines may represent an alternative to isolated islets for transplantation therapy of type 1 diabetes. Success of this approach may require development of cell lines that can withstand cytokine-mediated damage. To this end, we have cultured INS-1 insulinoma cells in increasing concentrations of interleukin-1beta (IL-1beta) + gamma-interferon (IFN-gamma), with approximate weekly iterations over an 8-week period. Based on the C,N diphenyl-N'-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium+ ++ bromide (MTT) viability assay, the selected cells, termed INS-1res, were 100% viable after 5 days of treatment with 10 ng/ml of IL-1beta. These cells were also 78 +/- 1.2% viable after 5 days of exposure to the combination of 10 ng/ml IL-1beta and 100 U/ml IFN-gamma, whereas parental INS-1 cells treated in the same manner were only 0.3 +/- 0.03% viable. INS-1res cells were also resistant to treatment with supernatants from activated rat peripheral blood mononuclear cells, whereas only 20% of parental INS-1 cells survived such treatment. The resistance to IL-1beta conferred by this procedure was stable, whereas the partial resistance to IFN-gamma was transient but reinducible by culture in the presence of cytokines. Stable transfection of INS-1res cells with a plasmid containing the human insulin cDNA and expansion of the transfected colonies in the absence of cytokines produced cell lines that were on average more resistant to IL-1beta + IFN-gamma (53 +/- 11%) than similarly transfected clones derived from parental INS-1 cells (15 +/- 7%). Importantly, several INS-1res-derived clones retained the capacity to secrete insulin in response to glucose concentrations over the normal physiological range. With regard to the mechanism by which selection was conferred, we found normal levels of IFN-gamma receptor mRNA, but a 60% reduction in expression of the IL-1 receptor type I (IL-1RI) in INS-1res cells compared with parental INS-1 cells. IL-1beta signaling through p38 MAP kinase was found to be normal in INS-1res cells, suggesting that their expression of IL-1RI is sufficient to maintain cytokine action. However, normal IL-1beta-mediated translocation of NF-kappaB and induction of inducible nitric oxide synthase expression and nitric oxide production was severely impaired in the INS-1res cell lines, suggesting a mechanism for the IL-1beta resistance. In sum, this study defines a strategy for isolation of cytokine-resistant beta-cell lines and provides a new system for studying the mechanisms by which such resistance can be achieved.


Subject(s)
Insulinoma , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Islets of Langerhans/cytology , Tumor Cells, Cultured , Animals , Cell Culture Techniques , Cell Death , Culture Media, Conditioned , Drug Resistance , Humans , Insulin/genetics , Islets of Langerhans/drug effects , Leukocytes, Mononuclear/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , RNA, Messenger/analysis , Rats , Receptors, Interferon/genetics , Receptors, Interleukin-1/genetics , Transfection , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects
19.
Diabetes ; 49(3): 424-30, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10868964

ABSTRACT

The biochemical mechanisms involved in regulation of insulin secretion are not completely understood. The rat INS-1 cell line has been used to gain insight in this area because it secretes insulin in response to glucose concentrations in the physiological range. However, the magnitude of the response is far less than that seen in freshly isolated rat islets. In the current study, we have stably transfected INS-1 cells with a plasmid containing the human proinsulin gene. After antibiotic selection and clonal expansion, 67% of the resultant clones were found to be poorly responsive to glucose in terms of insulin secretion (< or =2-fold stimulation by 15 mmol/l compared with 3 mmol/l glucose), 17% of the clones were moderately responsive (2- to 5-fold stimulation), and 16% were strongly responsive (5- to 13-fold stimulation). The differences in responsiveness could not be ascribed to differences in insulin content. Detailed analysis of one of the strongly responsive lines (832/13) revealed that its potent response to glucose (average of 10-fold) was stable over 66 population doublings (approximately 7.5 months of tissue culture) with half-maximal stimulation at 6 mmol/l glucose. Furthermore, in the presence of 15 mmol/l glucose, insulin secretion was potentiated significantly by 100 pmol/l isobutylmethylxanthine (320%), 1 mmol/l oleate/palmitate (77%), and 50 nmol/l glucagon-like peptide 1 (60%), whereas carbachol had no effect. Glucose-stimulated insulin secretion was also potentiated by the sulfonylurea tolbutamide (threefold at 3 mmol/l glucose and 50% at 15 mmol/l glucose) and was abolished by diazoxide, which demonstrates the operation of the ATP-sensitive K+ channel (K(ATP)) in 832/13 cells. Moreover, when the K(ATP) channel was bypassed by incubation of cells in depolarizing K+ (35 mmol/l), insulin secretion was more effectively stimulated by glucose in 832/13 cells than in parental INS-1 cells, which demonstrates the presence of a K(ATP) channel-independent pathway of glucose sensing. We conclude that clonal selection of INS-1 cells allows isolation of cell lines that exhibit markedly enhanced and stable responsiveness to glucose and several of its known potentiators. These lines may be attractive new vehicles for studies of beta-cell function.


Subject(s)
Adenosine Triphosphate/physiology , Glucose/pharmacology , Insulin/metabolism , Islets of Langerhans/metabolism , Potassium Channels/physiology , Animals , Cell Line , Clone Cells , Dose-Response Relationship, Drug , Drug Synergism , Humans , Insulin/genetics , Insulin Secretion , Proinsulin/metabolism , Protein Processing, Post-Translational , Rats , Transfection
20.
Am J Physiol Endocrinol Metab ; 278(3): E563-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10710512

ABSTRACT

Human obesity and high fat feeding in rats are associated with the development of insulin resistance and perturbed carbohydrate and lipid metabolism. It has been proposed that these metabolic abnormalities may be reversible by interventions that increase plasma leptin. Up to now, studies in nongenetic animal models of obesity and in human obesity have concentrated on multiple injection therapy with mixed results. Our study sought to determine whether a sustained, moderate increase in plasma leptin, achieved by administration of a recombinant adenovirus containing the leptin cDNA (AdCMV-leptin) would be effective in reversing the metabolic abnormalities of the obese phenotype. Wistar rats fed a high-fat diet (HF) were heavier (P < 0.05), had increased fat mass and intramuscular triglycerides (mTG), and had elevated plasma glucose, insulin, triglyceride, and free fatty acids compared with standard chow-fed (SC) control animals (all P < 0.01). HF rats also had impaired glucose tolerance and were markedly insulin resistant, as demonstrated by a 40% reduction in insulin-stimulated muscle glucose uptake (P < 0.001). Increasing plasma leptin levels to 29.0 +/- 1.5 ng/ml (from 7.0 +/- 1.4 ng/ml, P < 0.001) for a period of 6 days decreased adipose mass by 40% and normalized plasma glucose and insulin levels. In addition, insulin-stimulated skeletal muscle glucose uptake was normalized in hyperleptinemic rats, an effect that correlated closely with a 60% (P < 0.001) decrease in mTG. Importantly, HF rats that received a control adenovirus containing the beta-galactosidase cDNA and were calorically matched to AdCMV-leptin-treated animals remained hyperglycemic, hyperinsulinemic, insulin resistant, and maintained elevated mTG. We conclude that a gene-therapeutic intervention that elevates plasma leptin moderately for a sustained period reverses diet-induced hyperglycemia, hyperinsulinemia, and skeletal muscle insulin resistance, and that these improvements are tightly linked to leptin-induced reductions in mTG.


Subject(s)
Dietary Fats/adverse effects , Hyperglycemia/blood , Hyperinsulinism/blood , Insulin Resistance/physiology , Leptin/blood , Muscle, Skeletal/physiology , Animals , Blood/metabolism , Fasting/blood , Genetic Therapy , Hyperglycemia/etiology , Hyperglycemia/therapy , Hyperinsulinism/etiology , Hyperinsulinism/therapy , Leptin/genetics , Leptin/physiology , Male , Muscle, Skeletal/metabolism , Rats , Rats, Wistar , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...