Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Biochem J ; 359(Pt 3): 639-49, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11672439

ABSTRACT

We previously reported that SB203580, an inhibitor of p38 mitogen-activated protein kinase (p38 MAPK), attenuates insulin-stimulated glucose uptake without altering GLUT4 translocation. These results suggested that insulin might activate GLUT4 via a p38 MAPK-dependent pathway. Here we explore this hypothesis by temporal and kinetic analyses of the stimulation of GLUT4 translocation, glucose uptake and activation of p38 MAPK isoforms by insulin. In L6 myotubes stably expressing GLUT4 with an exofacial Myc epitope, we found that GLUT4 translocation (t(1/2)=2.5 min) preceded the stimulation of 2-deoxyglucose uptake (t(1/2)=6 min). This segregation of glucose uptake from GLUT4 translocation became more apparent when the two parameters were measured at 22 degrees C. Preincubation with the p38 MAPK inhibitors SB202190 and SB203580 reduced insulin-stimulated transport of either 2-deoxyglucose or 3-O-methylglucose by 40-60%. Pretreatment with SB203580 lowered the apparent transport V(max) of insulin-mediated 2-deoxyglucose and 3-O-methylglucose without any significant change in the apparent K(m) for either hexose. The IC(50) values for the partial inhibition of 2-deoxyglucose uptake by SB202190 and SB203580 were 1 and 2 microM respectively, and correlated with the IC(50) for full inhibition of p38 MAPK by the two inhibitors in myotubes (2 and 1.4 microM, respectively). Insulin caused a dose- (EC(50)=15 nM) and time- (t(1/2)=3 min) dependent increase in p38 MAPK phosphorylation, which peaked at 10 min (2.3+/-0.3-fold). In vitro kinase assay of immunoprecipitates from insulin-stimulated myotubes showed activation of p38 alpha (2.6+/-0.3-fold) and p38 beta (2.3+/-0.2-fold) MAPK. These results suggest that activation of GLUT4 follows GLUT4 translocation and that both mechanisms contribute to the full stimulation of glucose uptake by insulin. Furthermore, activation of GLUT4 may occur via an SB203580-sensitive pathway, possibly involving p38 MAPK.


Subject(s)
Glucose/metabolism , Insulin/metabolism , Mitogen-Activated Protein Kinases/metabolism , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Muscle, Skeletal/metabolism , Protein Serine-Threonine Kinases , Animals , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Glucose/analogs & derivatives , Glucose Transporter Type 4 , Humans , Isoenzymes , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Phosphorylation , Protein Transport/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , p38 Mitogen-Activated Protein Kinases
2.
J Biol Chem ; 276(49): 46079-87, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11598141

ABSTRACT

Phosphatidylinositol (PI) 3-kinase is required for insulin-stimulated translocation of GLUT4 to the surface of muscle and fat cells. Recent evidence suggests that the full stimulation of glucose uptake by insulin also requires activation of GLUT4, possibly via a p38 mitogen-activated protein kinase (p38 MAPK)-dependent pathway. Here we used L6 myotubes expressing Myc-tagged GLUT4 to examine at what level the signals regulating GLUT4 translocation and activation bifurcate. We compared the sensitivity of each process, as well as of signals leading to GLUT4 translocation (Akt and atypical protein kinase C) to PI 3-kinase inhibition. Wortmannin inhibited insulin-stimulated glucose uptake with an IC(50) of 3 nm. In contrast, GLUT4myc appearance at the cell surface was less sensitive to inhibition (IC(50) = 43 nm). This dissociation between insulin-stimulated glucose uptake and GLUT4myc translocation was not observed with LY294002 (IC(50) = 8 and 10 microm, respectively). The sensitivity of insulin-stimulated activation of PKC zeta/lambda, Akt1, Akt2, and Akt3 to wortmannin (IC(50) = 24, 30, 35, and 60 nm, respectively) correlated closely with inhibition of GLUT4 translocation. In contrast, insulin-dependent p38 MAPK phosphorylation was efficiently reduced in cells pretreated with wortmannin, with an IC(50) of 7 nm. Insulin-dependent p38 alpha and p38 beta MAPK activities were also markedly reduced by wortmannin (IC(50) = 6 and 2 nm, respectively). LY294002 or transient expression of a dominant inhibitory PI 3-kinase construct (Delta p85), however, did not affect p38 MAPK phosphorylation. These results uncover a striking correlation between PI 3-kinase, Akt, PKC zeta/lambda, and GLUT4 translocation on one hand and their segregation from glucose uptake and p38 MAPK activation on the other, based on their wortmannin sensitivity. We propose that a distinct, high affinity target of wortmannin, other than PI 3-kinase, may be necessary for activation of p38 MAPK and GLUT4 in response to insulin.


Subject(s)
Glucose/metabolism , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction , Androstadienes/pharmacology , Animals , Biological Transport , Cell Line , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Glucose Transporter Type 4 , Mitogen-Activated Protein Kinases/metabolism , Morpholines/pharmacology , Wortmannin , p38 Mitogen-Activated Protein Kinases
3.
Diabetes ; 50(6): 1464-71, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11375349

ABSTRACT

The cofactor of mitochondrial dehydrogenase complexes and potent antioxidant alpha-lipoic acid has been shown to lower blood glucose in diabetic animals. alpha-Lipoic acid enhances glucose uptake and GLUT1 and GLUT4 translocation in 3T3-L1 adipocytes and L6 myotubes, mimicking insulin action. In both cell types, insulin-stimulated glucose uptake is reduced by inhibitors of p38 mitogen-activated protein kinase (MAPK). Here we explore the effect of alpha-lipoic acid on p38 MAPK, phosphatidylinositol (PI) 3-kinase, and Akt1 in L6 myotubes. alpha-Lipoic acid (2.5 mmol/l) increased PI 3-kinase activity (31-fold) and Akt1 (4.9-fold). Both activities were inhibited by 100 nmol/l wortmannin. alpha-Lipoic acid also stimulated p38 MAPK phosphorylation by twofold within 10 min. The phosphorylation persisted for at least 30 min. Like insulin, alpha-lipoic acid increased the kinase activity of the alpha (2.8-fold) and beta (2.1-fold) isoforms of p38 MAPK, measured by an in vitro kinase assay. Treating cells with 10 micromol/l of the p38 MAPK inhibitors SB202190 or SB203580 reduced the alpha-lipoic acid-induced stimulation of glucose uptake by 66 and 55%, respectively. In contrast, SB202474, a structural analog that does not inhibit p38 MAPK, was without effect on glucose uptake. In contrast to 2-deoxyglucose uptake, translocation of GLUT4myc to the cell surface by either alpha-lipoic acid or insulin was unaffected by 20 micromol/l of SB202190 or SB203580. The results suggest that inhibition of 2-deoxyglucose uptake in response to alpha-lipoic acid by inhibitors of p38 MAPK is independent of an effect on GLUT4 translocation. Instead, it is likely that regulation of transporter activity is sensitive to these inhibitors.


Subject(s)
Arabidopsis Proteins , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Thioctic Acid/pharmacology , 3T3 Cells , Androstadienes/pharmacology , Animals , Biological Transport/drug effects , Deoxyglucose/antagonists & inhibitors , Deoxyglucose/pharmacokinetics , Enzyme Inhibitors/pharmacology , Glucose Transporter Type 4 , Imidazoles/pharmacology , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/physiology , Muscle Fibers, Skeletal/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Plant Proteins/metabolism , Potassium Channels/metabolism , Pyridines/pharmacology , Wortmannin , p38 Mitogen-Activated Protein Kinases
4.
Diabetes ; 49(11): 1794-800, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11078445

ABSTRACT

The stress-activated p38 mitogen-activated protein kinase (MAPK) was recently shown to be activated by insulin in muscle and adipose cells in culture. Here, we explore whether such stimulation is observed in rat skeletal muscle and whether muscle contraction can also affect the enzyme. Insulin injection (2 U over 3.5 min) resulted in increases in p38 MAPK phosphorylation measured in soleus (3.2-fold) and quadriceps (2.2-fold) muscles. Increased phosphorylation (3.5-fold) of an endogenous substrate of p38 MAPK, cAMP response element binder (CREB), was also observed. After in vivo insulin treatment, p38 MAPKalpha and p38 MAPKbeta isoforms were found to be activated (2.1- and 2.4-fold, respectively), using an in vitro kinase assay, in immunoprecipitates from quadriceps muscle extracts. In vitro insulin treatment (1 nmol/l over 4 min) and electrically-induced contraction of isolated extensor digitorum longus (EDL) muscle also doubled the kinase activity of p38 MAPKalpha and p38 MAPKbeta. The activity of both isoforms was inhibited in vitro by 10 micromol/l SB203580 in all muscles. To explore the possible participation of p38 MAPK in the stimulation of glucose uptake, EDL and soleus muscles were exposed to increasing doses of SB203580 before and during stimulation by insulin or contraction. SB203580 caused a significant reduction in the insulin- or contraction-stimulated 2-deoxyglucose uptake. Maximal inhibition (50-60%) occurred with 10 micromol/l SB203580. These results show that p38 MAPKalpha and -beta isoforms are activated by insulin and contraction in skeletal muscle. The data further suggest that activation of p38 MAPK may participate in the stimulation of glucose uptake by both stimuli in rat skeletal muscle.


Subject(s)
Insulin/pharmacology , Isoenzymes/metabolism , Mitogen-Activated Protein Kinases/metabolism , Muscle Contraction , Muscle, Skeletal/enzymology , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Deoxyglucose/metabolism , Electric Stimulation , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Immunosorbent Techniques , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscle, Skeletal/physiology , Phosphorylation , Pyridines/pharmacology , Rats , Rats, Wistar , p38 Mitogen-Activated Protein Kinases
5.
Mol Biol Cell ; 11(7): 2403-17, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10888677

ABSTRACT

Like neuronal synaptic vesicles, intracellular GLUT4-containing vesicles must dock and fuse with the plasma membrane, thereby facilitating insulin-regulated glucose uptake into muscle and fat cells. GLUT4 colocalizes in part with the vesicle SNAREs VAMP2 and VAMP3. In this study, we used a single-cell fluorescence-based assay to compare the functional involvement of VAMP2 and VAMP3 in GLUT4 translocation. Transient transfection of proteolytically active tetanus toxin light chain cleaved both VAMP2 and VAMP3 proteins in L6 myoblasts stably expressing exofacially myc-tagged GLUT4 protein and inhibited insulin-stimulated GLUT4 translocation. Tetanus toxin also caused accumulation of the remaining C-terminal VAMP2 and VAMP3 portions in Golgi elements. This behavior was exclusive to these proteins, because the localization of intracellular myc-tagged GLUT4 protein was not affected by the toxin. Upon cotransfection of tetanus toxin with individual vesicle SNARE constructs, only toxin-resistant VAMP2 rescued the inhibition of insulin-dependent GLUT4 translocation by tetanus toxin. Moreover, insulin caused a cortical actin filament reorganization in which GLUT4 and VAMP2, but not VAMP3, were clustered. We propose that VAMP2 is a resident protein of the insulin-sensitive GLUT4 compartment and that the integrity of this protein is required for GLUT4 vesicle incorporation into the cell surface in response to insulin.


Subject(s)
Insulin/metabolism , Membrane Proteins/metabolism , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Actins/metabolism , Animals , Biological Transport , Cell Line , Cell Membrane/metabolism , Glucose Transporter Type 4 , Insulin/pharmacology , Monosaccharide Transport Proteins/genetics , Muscle, Skeletal/cytology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , R-SNARE Proteins , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tetanus Toxin/metabolism , Vesicle-Associated Membrane Protein 3
6.
Diabetologia ; 43(3): 294-303, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10768090

ABSTRACT

AIMS/HYPOTHESIS: A natural cofactor of mitochondrial dehydrogenase complexes and a potent antioxidant, alpha-lipoic acid improves glucose metabolism in people with Type II (non-insulin-dependent) diabetes mellitus and in animal models of diabetes. In this study we investigated the cellular mechanism of action of alpha-lipoic acid in 3T3-L1 adipocytes. METHODS: We treated 3T3-L1 adipocytes with 2.5 mmol/l R (+) alpha-lipoic acid for 2 to 60 min, followed by assays of: 2-deoxyglucose uptake; glucose transporter 1 and 4 (GLUT1 and GLUT4) subcellular localization; tyrosine phosphorylation of the insulin receptor or of the insulin receptor substrate-1 in cell lysates; association of phosphatidylinositol 3-kinase activity with immunoprecipitates of proteins containing phosphotyrosine or of insulin receptor substrate-1 using a in vitro kinase assay; association of the p85 subunit of phosphatidylinositol 3-kinase with phosphotyrosine proteins or with insulin receptor substrate-1; and in vitro activity of immunoprecipitated Akt1. The effect of R (+) alpha-lipoic acid was also compared with that of S(-) alpha-lipoic acid. RESULTS: Short-term treatment of 3T3-L1 adipocytes with R (+) alpha-lipoic acid rapidly stimulated glucose uptake in a wortmannin-sensitive manner, induced a redistribution of GLUT1 and GLUT4 to the plasma membrane, caused tyrosine phosphorylation of insulin receptor substrate-1 and of the insulin receptor, increased the antiphosphotyrosine-associated and insulin receptor substrate-1 associated phosphatidylinositol 3-kinase activity and stimulated Akt activity. CONCLUSION/INTERPRETATION: These results indicate that R (+) alpha-lipoic acid directly activates lipid, tyrosine and serine/threonine kinases in target cells, which could lead to the stimulation of glucose uptake induced by this natural cofactor. These properties are unique among all agents currently used to lower glycaemia in animals and humans with diabetes.


Subject(s)
Adipocytes/metabolism , Glucose/metabolism , Insulin/pharmacology , Muscle Proteins , Protein Serine-Threonine Kinases , Thioctic Acid/pharmacology , 3T3 Cells , Adipocytes/drug effects , Androstadienes/pharmacology , Animals , Biological Transport/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Glucose Transporter Type 1 , Glucose Transporter Type 4 , Insulin Receptor Substrate Proteins , Mice , Monosaccharide Transport Proteins/metabolism , Osmolar Concentration , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptor, Insulin/metabolism , Tyrosine/metabolism , Wortmannin
7.
J Biol Chem ; 274(15): 10071-8, 1999 Apr 09.
Article in English | MEDLINE | ID: mdl-10187787

ABSTRACT

The precise mechanisms underlying insulin-stimulated glucose transport still require investigation. Here we assessed the effect of SB203580, an inhibitor of the p38 MAP kinase family, on insulin-stimulated glucose transport in 3T3-L1 adipocytes and L6 myotubes. We found that SB203580, but not its inactive analogue (SB202474), prevented insulin-stimulated glucose transport in both cell types with an IC50 similar to that for inhibition of p38 MAP kinase (0.6 microM). Basal glucose uptake was not affected. Moreover, SB203580 added only during the transport assay did not inhibit basal or insulin-stimulated transport. SB203580 did not inhibit insulin-stimulated translocation of the glucose transporters GLUT1 or GLUT4 in 3T3-L1 adipocytes as assessed by immunoblotting of subcellular fractions or by immunofluorescence of membrane lawns. L6 muscle cells expressing GLUT4 tagged on an extracellular domain with a Myc epitope (GLUT4myc) were used to assess the functional insertion of GLUT4 into the plasma membrane. SB203580 did not affect the insulin-induced gain in GLUT4myc exposure at the cell surface but largely reduced the stimulation of glucose uptake. SB203580 had no effect on insulin-dependent insulin receptor substrate-1 phosphorylation, association of the p85 subunit of phosphatidylinositol 3-kinase with insulin receptor substrate-1, nor on phosphatidylinositol 3-kinase, Akt1, Akt2, or Akt3 activities in 3T3-L1 adipocytes. In conclusion, in the presence of SB203580, insulin caused normal translocation and cell surface membrane insertion of glucose transporters without stimulating glucose transport. We propose that insulin stimulates two independent signals contributing to stimulation of glucose transport: phosphatidylinositol 3-kinase leads to glucose transporter translocation and a pathway involving p38 MAP kinase leads to activation of the recruited glucose transporter at the membrane.


Subject(s)
Adipocytes/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Glucose/metabolism , Imidazoles/pharmacology , Insulin/pharmacology , Mitogen-Activated Protein Kinases , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Muscles/metabolism , Nerve Tissue Proteins , Proto-Oncogene Proteins , Pyridines/pharmacology , 3-O-Methylglucose/metabolism , 3T3 Cells , Adipocytes/drug effects , Animals , Biological Transport/drug effects , Deoxyglucose/metabolism , Enzyme Inhibitors/chemistry , Glucose Transporter Type 1 , Glucose Transporter Type 3 , Glucose Transporter Type 4 , Imidazoles/chemistry , Mice , Muscles/drug effects , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt , Pyridines/chemistry , p38 Mitogen-Activated Protein Kinases
8.
Diabetologia ; 41(10): 1199-204, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9794108

ABSTRACT

Despite the important physiological role of insulin in the regulation of ionic homeostasis, primarily mediated by the Na+/K(+)-ATPase and Na+/K+/2Cl- cotransporter, the intracellular signalling molecules mediating this effect of insulin have not been elucidated. Treatment of 3T3-L1 fibroblasts with insulin increased total 86Rb+ (K+) uptake from 0.8 +/- 0.04 to 1.02 +/- 0.05 nmol.mg-1.protein-1.min-1 (p < 0.005). These changes in K+ flux, though small, can alter the membrane potential. Uptake occurred through both the Na+/K(+)-ATPase and Na+/K+/2Cl- cotransporter and both were stimulated by insulin. Interestingly, when bumetanide was used to inhibit the Na+/K+/2Cl- cotransporter prior to insulin action, no increase in 86Rb+ uptake via the Na+/K(+)-ATPase was observed. The structurally distinct phosphatidylinositol 3-kinase inhibitors wortmannin (50-200 nmol/l) and LY294002 (50 mumol/l) attenuated both total insulin-stimulated 86Rb+ uptake as well as uptake via the Na+/K(+)-ATPase and Na+/K+/2Cl- cotransporter. Neither the inhibitor of p70.S6 kinase activation, rapamycin (30 ng/ml) nor the mitogen activated protein kinase kinase inhibitor, PD098059 (50 mumol/l), had any effect on insulin's stimulation of K+ influx. A 10 mumol/l concentration of the protein kinase C (PKC) inhibitor bisindolylmaleimide attenuated insulin action but at 1 mumol/l it was ineffective, suggesting involvement of the atypical PKC-zeta isoform. We conclude that insulin-stimulated K+ uptake in 3T3-L1 fibroblasts appears to involve concerted regulation of both the Na+/K(+)-ATPase and Na+/K+/2Cl- cotransporter and we show for the first time that this process is signalled via a pathway involving phosphatidylinositol 3-kinase and PKC-zeta.


Subject(s)
3T3 Cells/drug effects , Insulin/pharmacology , Isoenzymes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Potassium/metabolism , Protein Kinase C/metabolism , 3T3 Cells/metabolism , Androstadienes/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Carrier Proteins/metabolism , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Isoenzymes/antagonists & inhibitors , Maleimides/pharmacology , Mice , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/antagonists & inhibitors , Ribosomal Protein S6 Kinases/antagonists & inhibitors , Rubidium Radioisotopes , Sirolimus/pharmacology , Sodium-Potassium-Chloride Symporters , Sodium-Potassium-Exchanging ATPase/metabolism , Wortmannin
9.
Biochem J ; 335 ( Pt 2): 351-6, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9761734

ABSTRACT

Insulin stimulates the rate of glucose transport into muscle and adipose cells by translocation of glucose transporter (GLUT4)-containing vesicles from an intracellular storage pool to the surface membrane. This event is mediated through the insulin receptor substrates (IRSs), which in turn activate phosphatidylinositol (PI) 3-kinase isoforms. It has been suggested that insulin causes attachment of PI 3-kinases to the intracellular GLUT4-containing vesicles in rat adipose cells. Furthermore, it has also been shown that GLUT4-containing vesicles in adipose cells contain a PI 4-kinase. In the present study we investigate whether GLUT4-containing vesicles isolated from rat skeletal muscle display PI 3-kinase and/or PI 4-kinase activities. Insulin stimulation caused a rapid increase (5-15-fold increase compared with control) in the intracellular cytosolic IRS-1-associated PI-3 kinase activity. This PI 3-kinase activity was also present in a membrane preparation containing the insulin-regulatable pool of GLUT4 transporters. However, when GLUT4-containing vesicles were isolated by immunoprecipitation from basal and insulin-stimulated (3 min) skeletal muscle, the vesicles displayed PI 4-kinase, but not PI 3-kinase, activity. Insulin did not regulate the PI 4-kinase activity in the GLUT4-containing vesicles. In conclusion, GLUT4-containing vesicles from rat skeletal muscle contain a PI 4-kinase, but not a PI 3-kinase. It is suggested that, in skeletal muscle, insulin causes activation of the IRS/PI 3-kinase complex in an intracellular membrane compartment associated closely with the GLUT4-containing vesicles, but not in the GLUT4-containing vesicles themselves.


Subject(s)
1-Phosphatidylinositol 4-Kinase/metabolism , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Muscle, Skeletal/metabolism , Phosphatidylinositol 3-Kinases/metabolism , 1-Phosphatidylinositol 4-Kinase/drug effects , Adenosine/pharmacology , Androstadienes/pharmacology , Animals , Cell Membrane/chemistry , Cell Membrane/metabolism , Glucose Transporter Type 4 , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Male , Monosaccharide Transport Proteins/analysis , Muscle, Skeletal/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Phosphoproteins/metabolism , Precipitin Tests , Rats , Rats, Wistar , Time Factors , Wortmannin
10.
Clin Ther ; 20(1): 125-40, 1998.
Article in English | MEDLINE | ID: mdl-9522110

ABSTRACT

The monomeric insulin analogue insulin lispro (Lys B28, Pro B29) is a rapid-acting insulin with a shorter duration of activity than human regular insulin. This compound has the advantage of reducing early postprandial hyperglycemia and the accompanying late hypoglycemia, thereby improving overall blood glucose control. To date, all published studies of the functional properties of insulin lispro have been conducted in whole animals. This study aimed to characterize the cellular actions of insulin lispro and the signals it elicits in an insulin-sensitive muscle cell line, L6 cells. Comparing the cellular actions of insulin lispro with those of human regular insulin, a number of observations were made. (1) Insulin lispro stimulated glucose and amino acid transport into L6 myotubes with a dose dependency and time course virtually identical to those of human regular insulin. (2) Insulin lispro was as effective as human regular insulin in stimulating time-dependent phosphorylation of insulin receptor substrate 1 (IRS-1), p70 ribosomal S6 kinase, and two isoforms of mitogen-activated protein kinase (ERK1 and ERK2). (3) Insulin lispro's ability to induce the association of IRS-1 with the p85 subunit of phosphatidylinositol 3-kinase was similar to that of human regular insulin. (4) As with human regular insulin, 100 nmol of the fungal metabolite wortmannin completely inhibited insulin lispro stimulation of glucose uptake. We concluded that the cellular actions of insulin lispro are similar to those of human regular insulin with respect to glucose and amino acid uptake and that the biochemical signals elicited are also comparable.


Subject(s)
Amino Acids/metabolism , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Insulin/analogs & derivatives , Insulin/physiology , Muscle, Skeletal/metabolism , Signal Transduction/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Deoxyglucose/metabolism , Humans , Insulin/pharmacology , Insulin Lispro , Insulin Receptor Substrate Proteins , Isoenzymes/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation
11.
Endocrinology ; 138(3): 1029-34, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9048605

ABSTRACT

vp165, a recently described member of the family of zinc-dependent membrane aminopeptidases, is a major constituent of glucose transporter-4 (GLUT4)-containing vesicles in adipocytes and skeletal muscle. Here we show that vp165 is expressed in L6 myoblasts and increases by 4.3-fold during differentiation into myotubes. The localization of vp165 in L6 myotubes was assessed by immunoblotting subcellular fractions from basal and insulin-stimulated cells and was compared to the distribution of GLUT4. vp165 and GLUT4 were mainly concentrated in the low density microsomal membranes under basal conditions. Upon stimulation with insulin, vp165 and GLUT4 were redistributed from the low density microsomes to the plasma membrane. The majority of vp165 was found in immunoisolated GLUT4-containing vesicles, and vice versa, the majority of GLUT4 was detected in immunoisolated vp165-containing vesicles. In contrast, the two other glucose transporter isoforms expressed in L6, GLUT1 and GLUT3, were excluded from GLUT4- and vp165-containing vesicles. These results suggest that in rat skeletal muscle cells, vp165 and GLUT4 cosegregate to the same intracellular compartment and that this is distinct from the compartment containing GLUT1 and GLUT3.


Subject(s)
Aminopeptidases/metabolism , Insulin/physiology , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Muscle, Skeletal/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Cystinyl Aminopeptidase , Glucose Transporter Type 4 , Microsomes/metabolism , Muscle, Skeletal/cytology , Rats , Subcellular Fractions/metabolism
12.
Diabetes ; 45(12): 1798-804, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8922368

ABSTRACT

Thioctic acid (alpha-lipoic acid), a natural cofactor in dehydrogenase complexes, is used in Germany in the treatment of symptoms of diabetic neuropathy. Thioctic acid improves insulin-responsive glucose utilization in rat muscle preparations and during insulin clamp studies performed in diabetic individuals. The aim of this study was to determine the direct effect of thioctic acid on glucose uptake and glucose transporters. In L6 muscle cells and 3T3-L1 adipocytes in culture, glucose uptake was rapidly increased by (R)-thioctic acid. The increment was higher than that elicited by the (S)-isomer or the racemic mixture and was comparable with that caused by insulin. In parallel to insulin action, the stimulation of glucose uptake by thioctic acid was abolished by wortmannin, an inhibitor of phosphatidylinositol 3-kinase, in both cell lines. Thioctic acid provoked an upward shift of the glucose-uptake insulin dose-response curve. The molar content of GLUT1 and GLUT4 transporters was measured in both cell lines. 3T3-L1 adipocytes were shown to have >10 times more glucose transporters but similar ratios of GLUT4:GLUT1 than L6 myotubes. The effect of (R)-thioctic acid on glucose transporters was studied in the L6 myotubes. Its stimulatory effect on glucose uptake was associated with an intracellular redistribution of GLUT1 and GLUT4 glucose transporters, similar to that caused by insulin, with minimal effects on GLUT3 transporters. In conclusion, thioctic acid stimulates basal glucose transport and has a positive effect on insulin-stimulated glucose uptake. The stimulatory effect is dependent on phosphatidylinositol 3-kinase activity and may be explained by a redistribution of glucose transporters. This is evidence that a physiologically relevant compound can stimulate glucose transport via the insulin signaling pathway.


Subject(s)
Glucose/metabolism , Insulin/metabolism , Muscle Proteins , Signal Transduction , Thioctic Acid/pharmacology , 3T3 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Androstadienes/pharmacology , Animals , Biological Transport/drug effects , Cell Line , Glucose Transporter Type 1 , Glucose Transporter Type 4 , Insulin/pharmacology , Insulin Antagonists/pharmacology , Mice , Monosaccharide Transport Proteins/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Rats , Wortmannin
13.
Diabetes ; 45(11): 1516-23, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8866555

ABSTRACT

Insulin-stimulated glucose uptake in skeletal muscle is mediated through the GLUT4 glucose transporter. Transgenic (TG) mice overexpressing human GLUT4 in skeletal muscle show an increased ability to handle a glucose load. Here, the participation of the overexpressed GLUT4 in the response to insulin was examined. In TG mouse muscle, the GLUT4 protein content was 10-fold higher in crude membrane (CM), sevenfold higher in internal membrane (IM), and 15-fold higher in a plasma membrane (PM)-rich fraction, relative to non-TG littermates. This suggested partial saturation of the normal sorting mechanisms. The distribution and abundance of the GLUT1 glucose transporter was not affected. Insulin injection (4.3 U/kg body wt) increased GLUT4 in the PM-rich fraction; the increase was threefold higher in TG than in non-TG mice. Insulin decreased the GLUT4 content of the IM in both animal groups and of a second, heavier intracellular membrane fraction only in TG mice. The net content of Na+-K+-pump subunits was 40-65% lower in CM from TG compared with non-TG littermates. In spite of this, insulin caused a three- to sixfold higher translocation of the alpha2 and beta1 subunits of the Na+-K+-pump in TG compared with non-TG animals. The results suggest that overexpression of GLUT4 confers to the muscle increased ability to translocate subunits of the Na+-K+-pump either as a direct consequence of the recruitment of glucose transporters or as an adaptation to the more demanding metabolic state.


Subject(s)
Insulin/pharmacology , Monosaccharide Transport Proteins/biosynthesis , Muscle Proteins , Muscle, Skeletal/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Blood Glucose/metabolism , Cell Membrane/metabolism , Gene Expression/drug effects , Glucose Clamp Technique , Glucose Transporter Type 4 , Humans , Macromolecular Substances , Mice , Mice, Transgenic , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Muscle, Skeletal/drug effects , Reference Values , Subcellular Fractions/metabolism
14.
Am J Physiol ; 270(5 Pt 1): C1421-9, 1996 May.
Article in English | MEDLINE | ID: mdl-8967443

ABSTRACT

We have previously shown that an acute insulin treatment induces redistribution of the alpha 2- and beta 1- isoforms of the Na+-K+-ATPase from intracellular membranes to plasma membranes detected on subcellular fractionation of mixed muscles and immunoblotting with isoform-specific antibodies (H. S. Hundal et al. J. Biol. Chem. 267: 5040-5043, 1992). In the present study we give both biochemical and morphological evidence that this insulin effect is operative in muscles composed mostly of oxidative (red) fibers but not in muscles composed mostly of glycolytic (white) fibers. The redistribution of the Na+-K+-ATPase alpha 2- and beta 1-isoforms after insulin injection was detected in membranes isolated from and muscles (soleus, red gastrocnemius, red rectus femoris, and red vastus lateralis) but not in membranes from white muscles (white gastrocnemius, tensor fasciae latae, white rectus femoris, and white vastus lateralis). After insulin injection, the potassium-dependent 3-O-methylfluorescein phosphatase activity of the enzyme was higher by 22% in the plasma membrane-enriched fraction and lower by 15% in the internal membrane fraction isolated from red but not from white muscles. Quantitative immunoelectron microscopy of ultrathin muscle cryosections showed that in vivo insulin stimulation augmented the density of Na+-K+-ATPase alpha 2- and beta 1- isoforms at the plasma membrane of soleus muscle by 80 and 124%, respectively, with no change in white gastrocnemius muscle. The effect of insulin to increase the content of Na+-K+-ATPase alpha 2- and beta 1-subunits in isolated plasma membranes was still observed when glycemia was prevented from dropping by using hyperinsulinemic-euglycemic clamps. We conclude that the insulin-induced redistribution of the alpha 2- and beta 1-isoforms of the Na+-K+-ATPase from an intracellular pool to the plasma membrane in restricted to oxidative fiber-type skeletal muscles. This may be related to the selective expression of beta 1-subunits in these fibers and implies that the beta 2-subunit, typical of glycolytic muscles, does not sustain translocation of alpha 2 beta 2-complexes.


Subject(s)
Insulin/pharmacology , Muscle Fibers, Skeletal/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Cell Membrane/metabolism , Glycolysis , Hyperinsulinism/metabolism , Male , Microscopy, Immunoelectron , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Reference Values , Sodium-Potassium-Exchanging ATPase/ultrastructure , Subcellular Fractions/metabolism
15.
Biochem Biophys Res Commun ; 213(2): 462-8, 1995 Aug 15.
Article in English | MEDLINE | ID: mdl-7646500

ABSTRACT

Syntaxins are a family of membrane proteins believed to participate in docking/fusing of arriving vesicles during membrane sorting and secretion. Of the six mammalian syntaxins known, only brain syntaxin 1A/1B has been biochemically characterized in its endogenous form. Syntaxin 4 mRNA is expressed in selective tissues including rat skeletal muscle, although it has not been studied at the protein level in any cell type. Therefore, we generated an affinity-purified antibody against syntaxin 4 to demonstrate that this 36 kDa protein is expressed in rat skeletal muscle and L6 muscle cells in culture. The content of the syntaxin 4 protein increased by 1.9-fold during differentiation of L6 myoblasts into myotubes. By subcellular fractionation the protein was mainly recovered in plasma membrane-enriched fractions of both red and white skeletal muscles and L6 myotubes. Coupled to the recent detection of vesicle associated membrane protein-2 and cellubrevin in skeletal muscle, syntaxin 4 may play a role in membrane traffic in this tissue.


Subject(s)
Membrane Proteins/analysis , Muscle, Skeletal/chemistry , Animals , Cell Differentiation , Cell Fractionation , Cell Line , Cell Membrane/chemistry , Gene Expression , Membrane Proteins/genetics , Muscle, Skeletal/ultrastructure , Qa-SNARE Proteins , RNA, Messenger/analysis , Rats , Syntaxin 1
16.
FEBS Lett ; 366(2-3): 109-14, 1995 Jun 12.
Article in English | MEDLINE | ID: mdl-7789525

ABSTRACT

The GLUT4 glucose transporter and the alpha 2 subunit of the Na+,K(+)-ATPase of rat skeletal muscle are two proteins which redistribute from intracellular membranes to plasma membranes following in vivo insulin stimulation. Here we show that although both proteins co-segregate after subcellular fractionation of unstimulated rat hindlimb muscles, they do not share the same intracellular residence inside the muscle fibre. By immunogold single- and double-labeling on ultrathin muscle cryosections with specific antibodies, the GLUT4 glucose transporter and the Na+,K(+)-ATPase alpha 2 subunit were observed on different vesicular structures within the cell. GLUT4 was detected on subsarcolemmal and perinuclear membranes, and at the junction between myofibrillar A and I bands where triads are localized. The alpha 2 subunit of the Na+,K(+)-ATPase was observed at the plasma membrane and in distinct subsarcolemmal vesicles and intermyofibrillar membranes. Quantitative analysis of double-labeling of GLUT4 and Na+,K(+)-ATPase alpha 2 subunit revealed that less than 6% of the two proteins co-localize in the same continuous vesicular structures. The differential intracellular localization of the two proteins was further confirmed by immunopurification of GLUT4-containing membranes from muscle homogenates, in which the alpha 2 subunit of the Na+,K(+)-ATPase was found only at the same extent as the alpha 1 subunit of the enzyme, a protein exclusively present at the plasma membrane.


Subject(s)
Escherichia coli Proteins , Glycoside Hydrolases , Intracellular Membranes/metabolism , Monosaccharide Transport Proteins/metabolism , Muscle Proteins/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Bacterial Proteins/chemistry , Glucose Transporter Type 4 , Glycosyltransferases/chemistry , Immunohistochemistry , Insulin/pharmacology , Male , Molecular Sequence Data , Muscles/drug effects , Rats , Rats, Sprague-Dawley , Subcellular Fractions/chemistry
17.
Endocrinology ; 136(6): 2505-12, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7750472

ABSTRACT

Many studies suggest that sulfonylureas (SUs) have direct extrapancreatic actions. The action of gliclazide, a new SU, was examined and compared to that of glyburide in L6 myotubes, a model of skeletal muscle. Gliclazide and glyburide increased 2-deoxy-D-glucose (2DG) uptake in a time- and dose-dependent fashion after 24 h to a maximum of 179% and 202% of the basal value, respectively (P < 0.001). Acute (30-min) insulin (10(-7) M) stimulated 2DG uptake to similar levels (203% of basal), but this effect was absent after maximum stimulation by SU. SU action did not require insulin and was not blocked by the protein synthesis inhibitor cycloheximide. To investigate the mechanism of stimulation of 2DG uptake, cells were fractionated, and total plasma membrane and internal membrane levels of glucose transporter (GLUT) isoforms were determined by immunoblotting. Both drugs significantly increased the total content (1.7-fold) and plasma membrane level (1.8-fold) of GLUT1, with no change in internal membrane. Total content and plasma membrane levels of GLUT4 and GLUT3 did not change or showed a small decrease. We conclude that the stimulation of glucose uptake in L6 cells by gliclazide and glyburide is associated not with a redistribution but, rather, with an increase in the total membrane content and plasma membrane level of GLUT1, which is independent of protein synthesis. These data suggest a novel action of SU to stabilize GLUT1 protein at the plasma membrane.


Subject(s)
Gliclazide/pharmacology , Glucose/metabolism , Glyburide/pharmacology , Monosaccharide Transport Proteins/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Animals , Biological Transport, Active/drug effects , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cycloheximide/pharmacology , Deoxyglucose/metabolism , Dose-Response Relationship, Drug , Gliclazide/administration & dosage , Glyburide/administration & dosage , Insulin/pharmacology , Kinetics , Rats , Tissue Distribution
18.
Biochem Biophys Res Commun ; 199(2): 662-70, 1994 Mar 15.
Article in English | MEDLINE | ID: mdl-8135807

ABSTRACT

Murine models of insulin resistance and diabetes are versatile and have been used to investigate genetic and metabolic disorders. However, the principal assays to assess insulin action, i.e., the euglycemic-hyperinsulinemic clamp and subcellular distribution of glucose transporters, have not been implemented in this species. Here we describe procedures which allow these methods to be adapted to mice. When normal C57bl/6j mice were infused with graded doses of insulin (1, 3, 10 or 30 mU/kg/min) during a euglycemic-hyerinsulinemic clamp, the glucose infusion rate necessary to maintain euglycemia increased in a dose-dependent manner (7.4 +/- 1.7, 13.1 +/- 3.6, 24.1 +/- 2.3 or 34.8 +/- 7.5 mg/kg/min), respectively. Hindlimb muscles were isolated and samples of 2-3 g were subjected to subcellular fractionation finalizing on 25%, 30% and 35% sucrose gradients. Fraction F25 (plasma membranes) was enriched in alpha 2 Na+/K(+)-ATPase and GLUT1 glucose transporters, whereas fraction F35 (intracellular membranes) was enriched in Ca(2+)-ATPase and GLUT4 glucose transporters. Following insulin treatment, GLUT4 increased in F25 and decreased in F35. Insulin treatment had no effect on GLUT1 in F25. However, unlike in rat skeletal muscle, GLUT1 was detectable in F35 and its content decreased in this fraction following insulin treatment. The results demonstrate that whole-body glucose utilization can be assessed in mice using euglycemic-hyperinsulinemic clamps and demonstrate how subcellular fractionation procedures can be applied to murine muscle. Murine muscle GLUT4 translocates from an intracellular storage site to the plasma membrane in response to insulin.


Subject(s)
Glucose/metabolism , Insulin/pharmacology , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Muscles/metabolism , Animals , Calcium-Transporting ATPases/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Glucose Clamp Technique , Glucose Transporter Type 1 , Glucose Transporter Type 4 , Hyperinsulinism/metabolism , Infusions, Intravenous , Insulin/administration & dosage , Insulin Resistance/physiology , Male , Mice , Mice, Inbred C57BL , Monosaccharide Transport Proteins/drug effects , Muscles/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism
20.
FEBS Lett ; 328(3): 253-8, 1993 Aug 16.
Article in English | MEDLINE | ID: mdl-8394248

ABSTRACT

Hindlimb skeletal muscles of the rat express two isoforms of the alpha (alpha 1 and alpha 2) and two isoforms of the beta (beta 1 and beta 2) subunits of the Na+,K(+)-ATPase. Because several muscles constitute the hindlimb, we investigated if specific isoforms are expressed in particular muscles. Northern blot analysis using isoform-specific cDNA probes demonstrated that soleus muscle expressed only the beta 1 transcript, whereas EDL or white gastrocnemius muscles expressed only the beta 2 transcript, and red gastrocnemius muscle expressed both mRNAs. All muscles tested expressed both alpha 1 and alpha 2 transcripts, albeit to various degrees: alpha 1 transcripts were present to about the same extent in all muscles but alpha 2 mRNA was 4-fold more abundant in soleus than in EDL for the same amount of total RNA. Beta subunit protein levels were investigated in purified plasma membrane fractions of pooled red (soleus + red gastrocnemius + red quadriceps) or white (white gastrocnemius + white quadriceps) muscles using isoform-specific antibodies. Red muscles expressed mostly the beta 1 protein while white muscles expressed mostly the beta 2 subunit. Both muscle groups had similar levels of alpha 1 or alpha 2 subunits, and crude membranes isolated from red muscles had 30% higher Na+,K(+)-ATPase activity than white muscle membranes. We conclude that oxidative muscles (slow and fast twitch) express beta 1 subunits, whereas glycolytic, fast twitch muscles express beta 2 subunits, and that both beta isoforms support the Na+,K(+)-ATPase activity of the alpha subunits.


Subject(s)
Muscles/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sodium-Potassium-Exchanging ATPase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...