Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
Add more filters










Publication year range
1.
Acta Clin Belg ; 68(2): 92-6, 2013.
Article in English | MEDLINE | ID: mdl-23967715

ABSTRACT

BACKGROUND: Few data exist documenting the survival experience of immigrated sub-Saharan patients infected by the human immunodeficiency virus (HIV) on renal replacement therapy (RRT). METHODS: This retrospective single centre pilot study includes 105 consecutive patients of sub-Saharan origin who started RRT in our unit, between January 1986 and April 2010. The aim was to analyse the characteristics and the survival rate on RRT of these patients. RESULTS: Out of 105 patients 81/105 (77%) were HIV-negative and 24/105(23%) were HIV-positive. HIV-positive patients were younger than HIV-negative patients and they were more often treated with peritoneal dialysis (PD) (21/24) than with haemodialysis (HD). Dialysis peritonitis was equally distributed between HIV-positive and HIV-negative patients. Because of opportunistic infections, duration of hospitalisation was longer for HIV-positive than for HIV-negative patients. In PD-treated patients, the number of hospitalisations tended to be greater in patients who experienced at least one peritonitis episode and the duration of hospitalisation also tended to be longer. The survival rate was better in patients younger than 50 years and in patients on HD, but was similar for both positive and negative HIV patients. CONCLUSIONS: To the best of our knowledge, these are the first data concerning patients who have emigrated from sub-Saharan Africa to Belgium, and who are on RTT. Their survival rate is better if they are younger than 50 years and on HD. As the majority of HIV patients were treated by PD in our center, a conclusion regarding survival on different dialysis modalities is not possible for this group of patients. Survival rates were similar for HIV-positive and HIV-negative patients despite longer duration of hospisalization for HIV-positive patients.


Subject(s)
HIV Infections/complications , Kidney Failure, Chronic/therapy , Renal Replacement Therapy , Adult , Africa South of the Sahara/ethnology , Belgium , Female , Humans , Male , Middle Aged , Pilot Projects , Prognosis , Proportional Hazards Models , Retrospective Studies , Statistics, Nonparametric , Survival Rate , Treatment Outcome
2.
Am J Physiol Endocrinol Metab ; 298(2): E179-92, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19887597

ABSTRACT

Activators of 5'-AMP-activated protein kinase (AMPK) 5-aminoimidazole-4-carboxamide-1-beta-d-ribofuranoside (AICAR), metformin, and exercise activate atypical protein kinase C (aPKC) and ERK and stimulate glucose transport in muscle by uncertain mechanisms. Here, in cultured L6 myotubes: AICAR- and metformin-induced activation of AMPK was required for activation of aPKC and ERK; aPKC activation involved and required phosphoinositide-dependent kinase 1 (PDK1) phosphorylation of Thr410-PKC-zeta; aPKC Thr410 phosphorylation and activation also required MEK1-dependent ERK; and glucose transport effects of AICAR and metformin were inhibited by expression of dominant-negative AMPK, kinase-inactive PDK1, MEK1 inhibitors, kinase-inactive PKC-zeta, and RNA interference (RNAi)-mediated knockdown of PKC-zeta. In mice, muscle-specific aPKC (PKC-lambda) depletion by conditional gene targeting impaired AICAR-stimulated glucose disposal and stimulatory effects of both AICAR and metformin on 2-deoxyglucose/glucose uptake in muscle in vivo and AICAR stimulation of 2-[(3)H]deoxyglucose uptake in isolated extensor digitorum longus muscle; however, AMPK activation was unimpaired. In marked contrast to AICAR and metformin, treadmill exercise-induced stimulation of 2-deoxyglucose/glucose uptake was not inhibited in aPKC-knockout mice. Finally, in intact rodents, AICAR and metformin activated aPKC in muscle, but not in liver, despite activating AMPK in both tissues. The findings demonstrate that in muscle AICAR and metformin activate aPKC via sequential activation of AMPK, ERK, and PDK1 and the AMPK/ERK/PDK1/aPKC pathway is required for metformin- and AICAR-stimulated increases in glucose transport. On the other hand, although aPKC is activated by treadmill exercise, this activation is not required for exercise-induced increases in glucose transport, and therefore may be a redundant mechanism.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Enzyme Activators/pharmacology , Glucose Transport Proteins, Facilitative/metabolism , Metformin/pharmacology , Muscle Fibers, Skeletal/drug effects , Protein Kinase C/metabolism , Ribonucleosides/pharmacology , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/pharmacology , Animals , Blood Glucose/metabolism , Enzyme Activation/drug effects , Enzyme Activation/physiology , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Glucose Transport Proteins, Facilitative/drug effects , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Physical Conditioning, Animal/physiology , Protein Kinase C/drug effects , Protein Kinase C/genetics , Rats , Second Messenger Systems/drug effects , Second Messenger Systems/physiology , Signal Transduction/drug effects , Signal Transduction/physiology
3.
Diabetologia ; 52(6): 1197-207, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19357831

ABSTRACT

AIMS/HYPOTHESIS: Previous findings in rodents used as a model of diabetes suggest that insulin activation of atypical protein kinase C (aPKC) is impaired in muscle, but, unexpectedly, conserved in liver, despite impaired hepatic protein kinase B (PKB/Akt) activation. Moreover, aPKC at least partly regulates two major transactivators: (1) hepatic sterol receptor binding protein-1c (SREBP-1c), which controls lipid synthesis; and (2) nuclear factor kappa B (NFkappaB), which promotes inflammation and systemic insulin resistance. METHODS: In Goto-Kakizaki rats used as a model of type 2 diabetes, we examined: (1) whether differences in hepatic aPKC and PKB activation reflect differences in activation of IRS-1- and IRS-2-dependent phosphatidylinositol 3-kinase (PI3K); (2) whether hepatic SREBP-1c and NFkappaB are excessively activated by aPKC; and (3) metabolic consequences of excessive activation of hepatic aPKC, SREBP-1c and NFkappaB. RESULTS: In liver, as well as in muscle, IRS-2/PI3K activation by insulin was intact, whereas IRS-1/PI3K activation by insulin was impaired. Moreover, hepatic IRS-2 is known to control hepatic aPKC during insulin activation. Against this background, selective inhibition of hepatic aPKC by adenoviral-mediated expression of mRNA encoding kinase-inactive aPKC or short hairpin RNA targeting Irs2 mRNA and partially depleting hepatic IRS-2 diminished hepatic SREBP-1c production and NFkappaB activities, concomitantly improving serum lipids and insulin signalling in muscle and liver. Similar improvements in SREBP-1c, NFkappaB and insulin signalling were seen in ob/ob mice following inhibition of hepatic aPKC. CONCLUSIONS/INTERPRETATION: In diabetic rodent liver, diminished PKB activation may largely reflect impaired IRS-1/PI3K activation, while conserved aPKC activation reflects retained IRS-2/PI3K activity. Hepatic aPKC may also contribute importantly to excessive SREPB-1c and NFkappaB activities. Excessive hepatic aPKC-dependent activation of SREBP-1c and NFkappaB may contribute importantly to hyperlipidaemia and systemic insulin resistance.


Subject(s)
Diabetes Mellitus/metabolism , Hyperlipidemias/metabolism , Insulin Resistance/physiology , Liver/metabolism , NF-kappa B/metabolism , Protein Kinase C/physiology , Sterol Regulatory Element Binding Protein 1/metabolism , Analysis of Variance , Animals , Blood Glucose/metabolism , Blotting, Western , Cholesterol/blood , Diabetes Mellitus/physiopathology , Disease Models, Animal , Electrophoretic Mobility Shift Assay , Hyperlipidemias/physiopathology , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Male , Muscles/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Rats , Rats, Wistar , Triglycerides/blood
4.
Diabetologia ; 52(1): 60-4, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18972094

ABSTRACT

AIMS/HYPOTHESES: Insulin-stimulated glucose transport in muscle is impaired in type 2 diabetes, presumably reflecting reduced activation of atypical protein kinase C (aPKC) and protein kinase B (PKB/Akt). As previously shown, reductions in aPKC activation are seen at sub-maximal and maximal insulin stimulation, reductions in PKB activation are best seen at sub-maximal insulin stimulation and aPKC reductions at maximal insulin are partly improved by thiazolidinedione or metformin treatment. However, effects of combined thiazolidinedione-metformin treatment on aPKC or PKB activation by sub-maximal and maximal insulin are unknown. METHODS: Type 2 diabetic patients were examined before and 5 to 6 weeks after combined thiazolidinedione-metformin therapy for activation of muscle aPKC and PKBbeta and their upstream activators, the insulin receptor (IR) and IRS-1-associated phosphatidylinositol 3-kinase (PI3K), during euglycaemic-hyperinsulinaemic clamp studies conducted with sub-maximal (400-500 pmol/l) and maximal (1400 pmol/l) insulin concentrations. RESULTS: Following combined thiazolidinedione-metformin therapy, increases in glucose disposal and increases in sub-maximal and maximal insulin-induced activities of all four muscle signalling factors, IR, IRS-1-dependent PI3K (IRS-1/PI3K), aPKC and PKBbeta, were observed. Increases in PKBbeta enzyme activity were accompanied by increases in phosphorylation of PKB and its substrate, AS160, which is needed for glucose transport. Despite improved aPKC activity, muscle aPKC levels, which are diminished in type 2 diabetes, were not altered. CONCLUSIONS/INTERPRETATION: Combined thiazolidinedione-metformin treatment markedly improves sub-maximal and maximal insulin signalling to IR, IRS-1/PI3K, aPKC and PKBbeta in type 2 diabetic muscle. These improvements exceed those previously reported after treatment with either agent alone.


Subject(s)
Diabetes Mellitus/metabolism , Insulin Receptor Substrate Proteins/metabolism , Metformin/pharmacology , Muscle, Skeletal/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Thiazolidinediones/pharmacology , Body Mass Index , Cholesterol/blood , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Diabetes Mellitus/blood , Diabetes Mellitus/enzymology , Female , Glucose Clamp Technique , Humans , Hypoglycemic Agents/pharmacology , Insulin Receptor Substrate Proteins/drug effects , Male , Middle Aged , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Triglycerides/blood
5.
Diabetologia ; 49(12): 3000-8, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17028898

ABSTRACT

AIMS/HYPOTHESIS: Insulin-stimulated glucose transport in muscle is impaired in obesity and type 2 diabetes, but alterations in levels of relevant signalling factors, i.e. atypical protein kinase C (aPKC) and protein kinase B (PKB/Akt), are still uncertain. Clamp studies using maximal insulin concentrations have revealed defects in activation of aPKC, but not PKB, in both obese non-diabetic and obese diabetic subjects. In contrast, clamp studies using submaximal insulin concentrations revealed defects in PKB activation/phosphorylation in obese non-diabetic and diabetic subjects, but changes in aPKC were not reported. The aim of this study was to test the hypothesis that dose-related effects of insulin may account for the reported differences in insulin signalling to PKB in diabetic muscle. SUBJECTS AND METHODS: We compared enzymatic activation of aPKC and PKB, and PKB phosphorylation (threonine-308 and serine-473) during hyperinsulinaemic-euglycaemic clamp studies using both submaximal (400-500 pmol/l) and maximal (1400 pmol/l) insulin levels in non-diabetic control and obese diabetic subjects. RESULTS: In lean control subjects, the submaximal insulin concentration increased aPKC activity and glucose disposal to approximately 50% of the maximal level and PKBbeta activity to 25% of the maximal level, but PKBalpha activity was not increased. In these subjects, phosphorylation of PKBalpha and PKBbeta was increased to near-maximal levels at submaximal insulin concentrations. In obese diabetic subjects, whereas aPKC activation was defective at submaximal and maximal insulin concentrations, PKBbeta activation and the phosphorylation of PKBbeta and PKBalpha were defective at submaximal, but not maximal, insulin concentrations. CONCLUSIONS/INTERPRETATIONS: Defective PKBbeta activation/phosphorylation, seen on submaximal insulin stimulation in diabetic muscle, may largely reflect impaired activation of insulin signalling factors present in concentrations greater than those needed for full PKB activation/phosphorylation. Defective aPKC activation, seen at all insulin levels, appears to reflect, at least partly, an impaired action of distal factors needed for aPKC activation, or poor aPKC responsiveness.


Subject(s)
Diabetes Mellitus, Type 2/enzymology , Insulin/pharmacology , Muscle, Skeletal/enzymology , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Biopsy , Blood Glucose/metabolism , C-Peptide/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Female , Glycated Hemoglobin/metabolism , Humans , Insulin/blood , Male , Middle Aged , Muscle, Skeletal/pathology , Obesity/blood , Obesity/complications , Obesity/enzymology , Protein Kinase C/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Triglycerides/blood
6.
Diabetologia ; 49(2): 375-82, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16395615

ABSTRACT

AIMS/HYPOTHESIS: Metformin is widely used for treating type 2 diabetes mellitus, but its actions are poorly understood. In addition to diminishing hepatic glucose output, metformin, in muscle, activates 5'-AMP-activated protein kinase (AMPK), which alone increases glucose uptake and glycolysis, diminishes lipid synthesis, and increases oxidation of fatty acids. Moreover, such lipid effects may improve insulin sensitivity and insulin-stimulated glucose uptake. Nevertheless, the effects of metformin on insulin-sensitive signalling factors in human muscle have only been partly characterised to date. Interestingly, other substances that activate AMPK, e.g., aminoimidazole-4-carboxamide-1-beta-D: -riboside (AICAR), simultaneously activate atypical protein kinase C (aPKC), which appears to be required for the glucose transport effects of AICAR and insulin. METHODS: Since aPKC activation is defective in type 2 diabetes, we evaluated effects of metformin therapy on aPKC activity in muscles of diabetic subjects during hyperinsulinaemic-euglycaemic clamp studies. RESULTS: After metformin therapy for 1 month, basal aPKC activity increased in muscle, with little or no change in insulin-stimulated aPKC activity. Metformin therapy for 8 to 12 months improved insulin-stimulated, as well as basal aPKC activity in muscle. In contrast, IRS-1-dependent phosphatidylinositol (PI) 3-kinase activity and Ser473 phosphorylation of protein kinase B were not altered by metformin therapy, whereas the responsiveness of muscle aPKC to PI-3,4,5-(PO(4))(3), the lipid product of PI 3-kinase, was improved. CONCLUSIONS/INTERPRETATION: These findings suggest that the activation of AMPK by metformin is accompanied by increases in aPKC activity and responsiveness in skeletal muscle, which may contribute to the therapeutic effects of metformin.


Subject(s)
Diabetes Mellitus, Type 2/enzymology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Metformin/pharmacology , Muscle, Skeletal/enzymology , Phosphatidylinositol Phosphates/pharmacology , Protein Kinase C/metabolism , AMP-Activated Protein Kinases , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/physiopathology , Enzyme Activation , Fatty Acids/metabolism , Female , Glucose Clamp Technique , Glycolysis , Humans , Hypoglycemic Agents/therapeutic use , Insulin Receptor Substrate Proteins , Male , Metformin/therapeutic use , Middle Aged , Multienzyme Complexes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
7.
Biochem Soc Trans ; 33(Pt 2): 350-3, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15787604

ABSTRACT

It now seems clear that aPKC (atypical protein kinase C) isoforms are required for insulin-stimulated glucose transport in muscle and adipocytes. Moreover, there are marked defects in the activation of aPKCs under a variety of insulin-resistant conditions in humans, monkeys and rodents. In humans, defects in aPKC in muscle are seen in Type II diabetes and its precursors, obesity, the obesity-associated polycystic ovary syndrome and impaired glucose tolerance. These defects in muscle aPKC activation are due to both impaired activation of insulin receptor substrate-1-dependent PI3K (phosphoinositide 3-kinase) and the direct activation of aPKCs by the lipid product of PI3K, PI-3,4,5-(PO4)3. Although it is still uncertain which underlying defect comes first, the resultant defect in aPKC activation in muscle most certainly contributes significantly to the development of skeletal muscle insulin resistance. Of further note, unlike the seemingly ubiquitous presence of defective aPKC activation in skeletal muscle in insulin-resistant states, the activation of aPKC is normal or increased in livers of Type II diabetic and obese rodents. The maintenance of aPKC activation in the liver may explain how insulin-dependent lipid synthesis is maintained in these states, as aPKCs function mainly in the activation of enzymes important for lipid synthesis. Thus increased activation of liver aPKC in hyperinsulinaemic states may contribute significantly to the development of hyperlipidaemia in insulin-resistant states.


Subject(s)
Insulin Resistance , Insulin/metabolism , Protein Kinase C/metabolism , Animals , Diabetes Mellitus/metabolism , Humans , Liver/metabolism , Muscles/metabolism
8.
J Clin Endocrinol Metab ; 89(8): 3994-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15292339

ABSTRACT

Insulin resistance in obesity is partly due to diminished glucose transport in myocytes and adipocytes, but underlying mechanisms are uncertain. Insulin-stimulated glucose transport requires activation of phosphatidylinositol (PI) 3-kinase (3K), operating downstream of insulin receptor substrate-1. PI3K stimulates glucose transport through increases in PI-3,4,5-(PO(4))(3) (PIP(3)), which activates atypical protein kinase C (aPKC) and protein kinase B (PKB/Akt). However, previous studies suggest that activation of aPKC, but not PKB, is impaired in intact muscles and cultured myocytes of obese subjects. Presently, we examined insulin activation of glucose transport and signaling factors in cultured adipocytes derived from preadipocytes harvested during elective liposuction in lean and obese women. Relative to adipocytes of lean women, insulin-stimulated [(3)H]2-deoxyglucose uptake and activation of insulin receptor substrate-1/PI3K and aPKCs, but not PKB, were diminished in adipocytes of obese women. Additionally, the direct activation of aPKCs by PIP(3) in vitro was diminished in aPKCs isolated from adipocytes of obese women. Similar impairment in aPKC activation by PIP(3) was observed in cultured myocytes of obese glucose-intolerant subjects. These findings suggest the presence of defects in PI3K and aPKC activation that persist in cultured cells and limit insulin-stimulated glucose transport in adipocytes and myocytes of obese subjects.


Subject(s)
Adipocytes/metabolism , Insulin/pharmacology , Muscle Fibers, Skeletal/metabolism , Obesity/metabolism , Phosphatidylinositol Phosphates/pharmacology , Protein Kinase C/metabolism , Stem Cells/cytology , Adult , Cells, Cultured , Deoxyglucose/pharmacokinetics , Enzyme Activation , Female , Humans , Insulin Receptor Substrate Proteins , Middle Aged , Obesity/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt
9.
J Biol Chem ; 276(38): 35537-45, 2001 Sep 21.
Article in English | MEDLINE | ID: mdl-11463795

ABSTRACT

Insulin controls glucose uptake by translocating GLUT4 and other glucose transporters to the plasma membrane in muscle and adipose tissues by a mechanism that appears to require protein kinase C (PKC)-zeta/lambda operating downstream of phosphatidylinositol 3-kinase. In diabetes mellitus, insulin-stimulated glucose uptake is diminished, but with hyperglycemia, uptake is maintained but by uncertain mechanisms. Presently, we found that glucose acutely activated PKC-zeta/lambda in rat adipocytes and rat skeletal muscle preparations by a mechanism that was independent of phosphatidylinositol 3-kinase but, interestingly, dependent on the apparently sequential activation of the dantrolene-sensitive, nonreceptor proline-rich tyrosine kinase-2; components of the extracellular signal-regulated kinase (ERK) pathway, including, GRB2, SOS, RAS, RAF, MEK1 and ERK1/2; and, most interestingly, phospholipase D, thus yielding increases in phosphatidic acid, a known activator of PKC-zeta/lambda. This activation of PKC-zeta/lambda, moreover, appeared to be required for glucose-induced increases in GLUT4 translocation and glucose transport in adipocytes and muscle cells. Our findings suggest the operation of a novel pathway for activating PKC-zeta/lambda and glucose transport.


Subject(s)
Glucose/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Muscle Proteins , Phospholipase D/metabolism , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , Adipocytes/enzymology , Adipocytes/metabolism , Androstadienes/pharmacology , Animals , Dantrolene/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Focal Adhesion Kinase 2 , Glucose Transporter Type 4 , Isoenzymes , Monosaccharide Transport Proteins/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Protein Transport , Rats , Wortmannin
10.
Endocrinology ; 142(4): 1595-605, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11250941

ABSTRACT

Atypical protein kinases C (PKCs), zeta and lambda, and protein kinase B (PKB) are thought to function downstream of phosphatidylinositol 3-kinase (PI 3-kinase) and regulate glucose transport during insulin action in skeletal muscle and adipocytes. Insulin-stimulated glucose transport is defective in type II diabetes mellitus, and this defect is ameliorated by thiazolidinediones and lowering of blood glucose by chronic insulin therapy or short-term fasting. Presently, we evaluated the effects of these insulin-sensitizing modalities on the activation of insulin receptor substrate-1 (IRS-1)-dependent PI 3-kinase, PKC-zeta/lambda, and PKB in vastus lateralis skeletal muscles and adipocytes of nondiabetic and Goto-Kakizaki (GK) diabetic rats. Insulin provoked rapid increases in the activity of PI 3-kinase, PKC-zeta/lambda, and PKB in muscles and adipocytes of nondiabetic rats, but increases in IRS-1-dependent PI 3-kinase and PKC-zeta/lambda, but not PKB, activity were substantially diminished in GK muscles and adipocytes. Rosiglitazone treatment for 10-14 days, 10-day insulin treatment, and 60-h fasting reversed defects in PKC-zeta/lambda activation in GK muscles and adipocytes and increased glucose transport in GK adipocytes, without necessarily increasing IRS-1-dependent PI 3-kinase or PKB activation. Our findings suggest that insulin-sensitizing modalities, viz. thiazolidinediones, chronic insulin treatment, and short-term fasting, similarly improve defects in insulin-stimulated glucose transport at least partly by correcting defects in insulin-induced activation of PKC-zeta/lambda.


Subject(s)
Adipocytes/enzymology , Fasting/metabolism , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Muscle, Skeletal/enzymology , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases , Thiazoles/pharmacology , Thiazolidinediones , Adipocytes/drug effects , Animals , Blotting, Western , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Enzyme Activators/pharmacology , Isoenzymes , Mice , Muscle, Skeletal/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Wistar , Rosiglitazone
11.
Biochemistry ; 40(1): 249-55, 2001 Jan 09.
Article in English | MEDLINE | ID: mdl-11141077

ABSTRACT

Activation of protein kinase C-zeta (PKC-zeta) by insulin requires phosphatidylinositol (PI) 3-kinase-dependent increases in phosphatidylinositol-3,4,5-(PO(4))(3) (PIP(3)) and phosphorylation of activation loop and autophosphorylation sites, but actual mechanisms are uncertain. Presently, we examined: (a) acute effects of insulin on threonine (T)-410 loop phosphorylation and (b) effects of (i) alanine (A) and glutamate (E) mutations at T410 loop and T560 autophosphorylation sites and (ii) N-terminal truncation on insulin-induced activation of PKC-zeta. Insulin acutely increased T410 loop phosphorylation, suggesting enhanced action of 3-phosphoinositide-dependent protein kinase-1 (PDK-1). Despite increasing in vitro autophosphorylation of wild-type PKC-zeta and T410E-PKC-zeta, insulin and PIP(3) did not stimulate autophosphorylation of T560A, T560E, T410A/T560E, T410E/T560A, or T410E/T560E mutant forms of PKC-zeta; thus, T560 appeared to be the sole autophosphorylation site. Activating effects of insulin and/or PIP(3) on enzyme activity were completely abolished in T410A-PKC-zeta, partially compromised in T560A-PKC-zeta, T410E/T560A-PKC-zeta, and T410A/T560E-PKC-zeta, and largely intact in T410E-PKC-zeta, T560E-PKC-zeta, and T410E/T560E-PKC-zeta. Activation of the T410E/T560E mutant suggested a phosphorylation-independent mechanism. As functional correlates, insulin effects on epitope-tagged GLUT4 translocation were compromised by expression of T410A-PKC-zeta, T560A-PKC-zeta, T410E/T560A, and T410A/T560E-PKC-zeta but not T410E-PKC-zeta, T560E-PKC-zeta, or T410E/T560E-PKC-zeta. Insulin, but not PIP(3), activated truncated, pseudosubstrate-lacking forms of PKC-zeta and PKC-lambda by a wortmannin-sensitive mechanism, apparently involving PI 3-kinase/PDK-1-dependent phosphorylations but independent of PIP(3)-dependent conformational activation. Our findings suggest that insulin, via PIP(3), provokes increases in PKC-zeta enzyme activity through (a) PDK-1-dependent T410 loop phosphorylation, (b) T560 autophosphorylation, and (c) phosphorylation-independent/conformational-dependent relief of pseudosubstrate autoinhibition.


Subject(s)
Insulin/pharmacology , Phosphatidylinositol Phosphates/physiology , Protein Kinase C/metabolism , Threonine/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Adipocytes/enzymology , Amino Acid Substitution/genetics , Animals , Enzyme Activation/genetics , Glutamic Acid/genetics , Insulin/chemistry , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/chemistry , Phosphorylation , Protein Kinase C/biosynthesis , Protein Kinase C/genetics , Protein Serine-Threonine Kinases/physiology , Protein Structure, Secondary , Protein Structure, Tertiary/genetics , Rats , Recombinant Proteins/pharmacology , Sequence Deletion , Substrate Specificity/genetics , Threonine/biosynthesis , Threonine/genetics , Transfection
12.
Endocrinology ; 141(11): 4120-7, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11089544

ABSTRACT

We used adenoviral gene transfer methods to evaluate the role of atypical protein kinase Cs (PKCs) during insulin stimulation of glucose transport in L6 myotubes. Expression of wild-type PKC-lambda potentiated maximal and half-maximal effects of insulin on 2-deoxyglucose uptake, but did not alter basal uptake. Expression of constitutively active PKC-lambda enhanced basal 2-deoxyglucose uptake to virtually the same extent as that observed during insulin treatment. In contrast, expression of kinase-defective PKC-lambda completely blocked insulin-stimulated, but not basal, 2-deoxyglucose uptake. Similar to alterations in glucose transport, constitutively active PKC-lambda mimicked, and kinase-defective PKC-lambda completely inhibited, insulin effects on GLUT4 glucose transporter translocation to the plasma membrane. Expression of kinase-defective PKC-lambda, in addition to inhibition of atypical PKC enzyme activity, was attended by paradoxical increases in GLUT4 and GLUT1 glucose transporter levels and insulin-stimulated protein kinase B enzyme activity. Our findings suggest that in L6 myotubes, 1) atypical PKCs are required and sufficient for insulin-stimulated GLUT4 translocation and glucose transport; and 2) activation of protein kinase B in the absence of activation of atypical PKCs is insufficient for insulin-induced activation of glucose transport.


Subject(s)
Adenoviridae/genetics , Gene Transfer, Horizontal , Glucose/metabolism , Insulin/pharmacology , Muscle Proteins , Muscle, Skeletal/metabolism , Protein Kinase C/genetics , Protein Serine-Threonine Kinases , Animals , Biological Transport/drug effects , Cell Line , Cell Membrane/metabolism , Gene Expression , Glucose Transporter Type 1 , Glucose Transporter Type 4 , Isoenzymes/genetics , Isoenzymes/metabolism , Monosaccharide Transport Proteins/metabolism , Mutation , Protein Kinase C/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats
13.
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
14.
J Biol Chem ; 275(22): 16690-6, 2000 Jun 02.
Article in English | MEDLINE | ID: mdl-10749857

ABSTRACT

We evaluated effects of the thiazolidinedione, rosiglitazone, on insulin-induced activation of protein kinase C (PKC)-zeta/lambda and glucose transport in adipocytes of Goto-Kakizaki (GK)-diabetic and nondiabetic rats. Insulin effects on PKC-zeta/lambda and 2-deoxyglucose uptake were diminished by approximately 50% in GK adipocytes, as compared with control adipocytes. This defect in insulin-induced PKC-zeta/lambda activation was associated with diminished activation of IRS-1-dependent phosphatidylinositol (PI) 3-kinase, and was accompanied by diminished phosphorylation of threonine 410 in the activation loop of PKC-zeta; in contrast, protein kinase B (PKB) activation and phosphorylation were not significantly altered. Rosiglitazone completely reversed defects in insulin-stimulated 2-deoxyglucose uptake, PKCzeta/lambda enzyme activity and PKC-zeta threonine 410 phosphorylation, but had no effect on PI 3-kinase activation or PKB activation/phosphorylation in GK adipocytes. Similarly, in adipocytes of nondiabetic rats, rosiglitazone provoked increases in insulin-stimulated 2-deoxyglucose uptake, PKC-zeta/lambda enzyme activity and phosphorylation of both threonine 410 activation loop and threonine 560 autophosphorylation sites in PKC-zeta, but had no effect on PI 3-kinase activation or PKB activation/phosphorylation. Our findings suggest that (a) decreased effects of insulin on glucose transport in adipocytes of GK-diabetic rats are due at least in part to diminished phosphorylation/activation of PKC-zeta/lambda, and (b) thiazolidinediones enhance glucose transport responses to insulin in adipocytes of both diabetic and nondiabetic rats through increases in phosphorylation/activation of PKC-zeta/lambda.


Subject(s)
Adipocytes/drug effects , Diabetes Mellitus, Experimental/metabolism , Glucose/metabolism , Insulin/physiology , Isoenzymes/metabolism , Protein Kinase C/metabolism , Thiazoles/pharmacology , Thiazolidinediones , Adipocytes/metabolism , Animals , Biological Transport , Diabetes Mellitus, Experimental/enzymology , Enzyme Activation , Male , Rats , Rats, Wistar
15.
J Biol Chem ; 274(43): 30495-500, 1999 Oct 22.
Article in English | MEDLINE | ID: mdl-10521430

ABSTRACT

The mechanisms used by insulin to activate the multifunctional intracellular effectors, extracellular signal-regulated kinases 1 and 2 (ERK1/2), are only partly understood and appear to vary in different cell types. Presently, in rat adipocytes, we found that insulin-induced activation of ERK was blocked (a) by chemical inhibitors of both phosphatidylinositol 3-kinase (PI3K) and protein kinase C (PKC)-zeta, and, moreover, (b) by transient expression of both dominant-negative Deltap85 PI3K subunit and kinase-inactive PKC-zeta. Further, insulin effects on ERK were inhibited by kinase-inactive 3-phosphoinositide-dependent protein kinase-1 (PDK-1), and by mutation of Thr-410 in the activation loop of PKC-zeta, which is the target of PDK-1 and is essential for PI3K/PDK-1-dependent activation of PKC-zeta. In addition to requirements for PI3K, PDK-1, and PKC-zeta, we found that a tyrosine kinase (presumably the insulin receptor), the SH2 domain of GRB2, SOS, RAS, RAF, and MEK1 were required for insulin effects on ERK in the rat adipocyte. Our findings therefore suggested that PDK-1 and PKC-zeta serve as a downstream effectors of PI3K, and act in conjunction with GRB2, SOS, RAS, and RAF, to activate MEK and ERK during insulin action in rat adipocytes.


Subject(s)
Adipocytes/enzymology , Insulin/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Amino Acid Substitution , Androstadienes/pharmacology , Animals , Chromones/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epididymis , Flavonoids/pharmacology , Genistein/pharmacology , Kinetics , Male , Mitogen-Activated Protein Kinase 3 , Morpholines/pharmacology , Mutagenesis, Site-Directed , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/chemistry , Rats , Rats, Sprague-Dawley , Receptor, Insulin/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Threonine , Wortmannin , src Homology Domains
16.
Mol Endocrinol ; 13(10): 1766-72, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10517677

ABSTRACT

Previous studies have suggested that 1) atypical protein kinase C (PKC) isoforms are required for insulin stimulation of glucose transport, and 2) 3-phosphoinositide-dependent protein kinase-1 (PDK-1) is required for activation of atypical PKCs. Presently, we evaluated the role of PDK-1, both in the activation of PKC-zeta, and the translocation of epitope-tagged glucose transporter 4 (GLUT4) to the plasma membrane, during insulin action in transiently transfected rat adipocytes. Overexpression of wild-type PDK-1 provoked increases in the activity of cotransfected hemagglutinin (HA)-tagged PKC-zeta and concomitantly enhanced HA-tagged GLUT4 translocation. Expression of both kinase-inactive PDK-1 and an activation-resistant form of PKC-zeta that is mutated at Thr-410, the immediate target of PDK-1 in the activation loop of PKC-zeta, inhibited insulin-induced increases in both HA-PKC-zeta activity and HA-GLUT4 translocation to the same extent as kinase-inactive PKC-zeta. Moreover, the inhibitory effects of kinase-inactive PDK-1 were fully reversed by cotransfection of wild-type PDK-1 and partly reversed by wild-type PKC-zeta, but not by wild-type PKB. In contrast to the T410A PKC-zeta mutant, an analogous double mutant of PKB (T308A/S473A) that is resistant to PDK-1 activation had only a small effect on insulin-stimulated HA-GLUT4 translocation and did not inhibit HA-GLUT4 translocation induced by overexpression of wild-type PDK-1. Our findings suggest that both PDK-1 and its downstream target, Thr-410 in the activation loop of PKC-zeta, are required for insulin-stimulated glucose transport.


Subject(s)
Insulin/metabolism , Isoenzymes/metabolism , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Biological Transport , Enzyme Activation , Epitopes/metabolism , Gene Expression Regulation , Glucose Transporter Type 4 , Hemagglutinins/genetics , Hemagglutinins/metabolism , Insulin/pharmacology , Isoenzymes/drug effects , Isoenzymes/genetics , Monosaccharide Transport Proteins/drug effects , Monosaccharide Transport Proteins/genetics , Mutation , Phosphorylation , Protein Kinase C/drug effects , Protein Kinase C/genetics , Protein Kinase C-theta , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Threonine/metabolism , Transfection
17.
Endocrinology ; 140(10): 4470-7, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10499500

ABSTRACT

The beta-isoform of protein kinase C (PKC) has paradoxically been suggested to be important for both insulin action and insulin resistance as well as for contributing to the pathogenesis of diabetic complications. Presently, we evaluated the effects of knockout of the PKCbeta gene on overall glucose homeostasis and insulin regulation of glucose transport. To evaluate subtle differences in glucose homeostasis in vivo, knockout mice were extensively backcrossed in C57BL/6 mice to diminish genetic differences other than the absence of the PKCbeta gene. PKCbeta-/- knockout offspring obtained through this backcrossing had 10% lower blood glucose levels than those observed in PKCbeta+/+ wild-type offspring in both the fasting state and 30 min after i.p. injection of glucose despite having similar or slightly lower serum insulin levels. Also, compared with commercially obtained C57BL/6-129/SV hybrid control mice, serum glucose levels were similar, and serum insulin levels were similar or slightly lower, in C57BL/6-129/SV hybrid PKCbeta knockout mice in fasting and fed states and after i.p. glucose administration. In keeping with a tendency for slightly lower serum glucose and/or insulin levels in PKCbeta knockout mice, insulin-stimulated 2-deoxyglucose (2-DOG) uptake was enhanced by 50-100% in isolated adipocytes; basal and insulin-stimulated epitope-tagged GLUT4 translocations in adipocytes were increased by 41% and 27%, respectively; and basal 2-DOG uptake was mildly increased by 20-25% in soleus muscles incubated in vitro. The reason for increased 2-DOG uptake and/or GLUT4 translocation in these tissues was uncertain, as there were no significant alterations in phosphatidylinositol 3-kinase activity or activation or in levels of GLUT1 or GLUT4 glucose transporters or other PKC isoforms. On the other hand, increases in 2-DOG uptake may have been partly caused by the loss of PKCbeta1, rather than PKCbeta2, as transient expression of PKCbeta1 selectively inhibited insulin-stimulated translocation of epitope-tagged GLUT4 in adipocytes prepared from PKCbeta knockout mice. Our findings suggest that 1) PKCbeta is not required for insulin-stimulated glucose transport; 2) overall glucose homeostasis in vivo is mildly enhanced by knockout of the PKCbeta gene; 3) glucose transport is increased in some tissues in PKCbeta knockout mice; and 4) increased glucose transport may be partly due to loss of PKCbeta1, which negatively modulates insulin-stimulated GLUT4 translocation.


Subject(s)
Glucose/metabolism , Homeostasis , Isoenzymes/genetics , Mice, Knockout/genetics , Mice, Knockout/metabolism , Muscle Proteins , Protein Kinase C/genetics , Adipocytes/metabolism , Animals , Biological Transport , Blood Glucose/analysis , Deoxyglucose/pharmacokinetics , Glucose Transporter Type 4 , Heterozygote , Insulin/blood , Insulin/pharmacology , Male , Mice , Mice, Inbred Strains , Monosaccharide Transport Proteins/metabolism , Muscle, Skeletal/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C beta
18.
J Biol Chem ; 274(36): 25308-16, 1999 Sep 03.
Article in English | MEDLINE | ID: mdl-10464256

ABSTRACT

In rat adipocytes, insulin provoked rapid increases in (a) endogenous immunoprecipitable combined protein kinase C (PKC)-zeta/lambda activity in plasma membranes and microsomes and (b) immunoreactive PKC-zeta and PKC-lambda in GLUT4 vesicles. Activity and autophosphorylation of immunoprecipitable epitope-tagged PKC-zeta and PKC-lambda were also increased by insulin in situ and phosphatidylinositol 3,4,5-(PO(4))(3) (PIP(3)) in vitro. Because phosphoinositide-dependent kinase-1 (PDK-1) is required for phosphorylation of activation loops of PKC-zeta and protein kinase B, we compared their activation. Both RO 31-8220 and myristoylated PKC-zeta pseudosubstrate blocked insulin-induced activation and autophosphorylation of PKC-zeta/lambda but did not inhibit PDK-1-dependent (a) protein kinase B phosphorylation/activation or (b) threonine 410 phosphorylation in the activation loop of PKC-zeta. Also, insulin in situ and PIP(3) in vitro activated and stimulated autophosphorylation of a PKC-zeta mutant, in which threonine 410 is replaced by glutamate (but not by an inactivating alanine) and cannot be activated by PDK-1. Surprisingly, insulin activated a truncated PKC-zeta that lacks the regulatory (presumably PIP(3)-binding) domain; this may reflect PIP(3) effects on PDK-1 or transphosphorylation by endogenous full-length PKC-zeta. Our findings suggest that insulin activates both PKC-zeta and PKC-lambda in plasma membranes, microsomes, and GLUT4 vesicles by a mechanism requiring increases in PIP(3), PDK-1-dependent phosphorylation of activation loop sites in PKC-zeta and lambda, and subsequent autophosphorylation and/or transphosphorylation.


Subject(s)
Adipocytes/metabolism , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Protein Kinase C/metabolism , Signal Transduction/drug effects , Adipocytes/ultrastructure , Animals , Biological Transport/drug effects , Cell Line , Cell Membrane/metabolism , Cytoplasmic Granules/metabolism , Glucose Transporter Type 4 , Isoenzymes , Mice , Phosphorylation , Rats
19.
J Biol Chem ; 274(20): 14074-8, 1999 May 14.
Article in English | MEDLINE | ID: mdl-10318822

ABSTRACT

Okadaic acid, an inhibitor of protein phosphatases 1 and 2A, is known to provoke insulin-like effects on GLUT4 translocation and glucose transport, but the underlying mechanism is obscure. Presently, we found in both rat adipocytes and 3T3/L1 adipocytes that okadaic acid provoked partial insulin-like increases in glucose transport, which were inhibited by phosphatidylinositol (PI) 3-kinase inhibitors, wortmannin and LY294002, and inhibitors of atypical protein kinase C (PKC) isoforms, zeta and lambda. Moreover, in both cell types, okadaic acid provoked increases in the activity of immunoprecipitable PKC-zeta/lambda by a PI 3-kinase-dependent mechanism. In keeping with apparent PI 3-kinase dependence of stimulatory effects of okadaic acid on glucose transport and PKC-zeta/lambda activity, okadaic acid provoked insulin-like increases in membrane PI 3-kinase activity in rat adipocytes; the mechanism for PI 3-kinase activation was uncertain, however, because it was not apparent in phosphotyrosine immunoprecipitates. Of further note, okadaic acid provoked partial insulin-like increases in the translocation of hemagglutinin antigen-tagged GLUT4 to the plasma membrane in transiently transfected rat adipocytes, and these stimulatory effects on hemagglutinin antigen-tagged GLUT4 translocation were inhibited by co-expression of kinase-inactive forms of PKC-zeta and PKC-lambda but not by a double mutant (T308A, S473A), activation-resistant form of protein kinase B. Our findings suggest that, as with insulin, PI 3-kinase-dependent atypical PKCs, zeta and lambda, are required for okadaic acid-induced increases in GLUT4 translocation and glucose transport in rat adipocytes and 3T3/L1 adipocytes.


Subject(s)
Adipocytes/enzymology , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Okadaic Acid/pharmacology , Protein Kinase C/metabolism , 3T3 Cells , Adipocytes/drug effects , Androstadienes/pharmacology , Animals , Deoxyglucose/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Glucose Transporter Type 4 , Indoles/pharmacology , Isoenzymes , Mice , Phosphatidylinositol 3-Kinases/metabolism , Platelet Aggregation Inhibitors/pharmacology , Protein Kinase C/antagonists & inhibitors , Rats , Wortmannin
20.
Endocrinology ; 140(5): 2145-51, 1999 May.
Article in English | MEDLINE | ID: mdl-10218965

ABSTRACT

The activation of c-Jun N-terminal kinase (JNK) by insulin and anisomycin has been reported to result in increases in glycogen synthase (GS) activity in rat skeletal muscle (Moxham et al., J Biol Chem, 1996, 271:30765-30773). In addition, the protein kinase C (PKC) inhibitor, RO 31-8220, has been reported to activate JNK in rat-1 fibroblasts (Beltman et al., J Biol Chem, 1996, 271:27018-27024). Presently, we found that the RO 31-8220, as well as insulin, activated JNK and GS and stimulated glucose incorporation into glycogen in rat adipocytes and L6 myotubes. In contrast to activation of JNK, RO 31-8220 inhibited extracellular response kinases 1 and 2 (ERK1/2) and had no significant effects on protein kinase B (PKB). Stimulatory effects of RO 31-8220 on JNK and glycogen metabolism were not explained by PKC inhibition, as other PKC inhibitors were without effect on glucose incorporation into glycogen and have no effect on JNK (Beltman et al., J Biol Chem, 1996, 271:27018). Insulin, on the other hand, activated JNK, as well as PKB and ERK1/2. However, stimulatory effects of insulin on GS and glucose incorporation into glycogen appeared to be fully intact and additive to those of RO 31-8220, despite the fact that insulin did not provoke additive increases in JNK activity above those observed with RO 31-8220 alone. Our findings suggest that JNK serves to activate GS during the action of RO 31-8220 in rat adipocytes and L6 myotubes; insulin, on the other hand, appears to activate GS largely independently of JNK.


Subject(s)
Adipocytes/enzymology , Enzyme Inhibitors/pharmacology , Glycogen Synthase/metabolism , Indoles/pharmacology , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Muscles/enzymology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Adipocytes/drug effects , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Enzyme Activation/drug effects , Glycogen/metabolism , Insulin/pharmacology , MAP Kinase Kinase 4 , Male , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Muscles/drug effects , Protein Kinase C/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...