Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Cardiovasc Transl Res ; 11(3): 236-245, 2018 06.
Article in English | MEDLINE | ID: mdl-29392537

ABSTRACT

Gene-targeted therapy with the inotropic Ca2 + -sensor protein S100A1 rescues contractile function in post-ischemic heart failure and is being developed towards clinical trials. Its proven beneficial effect on cardiac metabolism and mitochondrial function suggests a cardioprotective effect of S100A1 in myocardial ischemia-reperfusion injury (IRI). Fivefold cardiomyocyte-specific S100A1 overexpressing, isolated rat hearts perfused in working mode were subjected to 28 min ischemia (37 °C) followed by 60 min reperfusion. S100A1 overexpressing hearts showed superior hemodynamic recover: Left ventricular pressure recovered to 57 ± 7.3% of baseline compared to 51 ± 4.6% in control (p = 0.025), this effect mirrored in LV work and dP/dt(max). Troponin T and lactate dehydrogenase was decreased in the S100A1 group, as well as FoxO pro-apoptotic transcription factor, indicating less tissue necrosis, whereas phosphocreatine content was higher after reperfusion. This is the first report of a cardioprotective effect of S100A1 overexpression in a global IRI model.


Subject(s)
Genetic Therapy/methods , Myocardial Contraction , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , S100 Proteins/biosynthesis , Ventricular Function, Left , Animals , Dependovirus , Disease Models, Animal , Gene Transfer Techniques , Genetic Vectors , Humans , Isolated Heart Preparation , Male , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Rats, Inbred Lew , Recovery of Function , S100 Proteins/genetics , Up-Regulation , Ventricular Pressure
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 ; 48(12): 2591-601, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16283248

ABSTRACT

AIMS/HYPOTHESIS: 5'AMP-activated protein kinase (AMPK) and insulin stimulate glucose transport in heart and muscle. AMPK acts in an additive manner with insulin to increase glucose uptake, thereby suggesting that AMPK activation may be a useful strategy for ameliorating glucose uptake, especially in cases of insulin resistance. In order to characterise interactions between the insulin- and AMPK-signalling pathways, we investigated the effects of AMPK activation on insulin signalling in the rat heart in vivo. METHODS: Male rats (350-400 g) were injected with 1 g/kg 5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside (AICAR) or 250 mg/kg metformin in order to activate AMPK. Rats were administered insulin 30 min later and after another 30 min their hearts were removed. The activities and phosphorylation levels of components of the insulin-signalling pathway were subsequently analysed in individual rat hearts. RESULTS: AICAR and metformin administration activated AMPK and enhanced insulin signalling downstream of protein kinase B in rat hearts in vivo. Insulin-induced phosphorylation of glycogen synthase kinase 3 (GSK3) beta, p70 S6 kinase (p70S6K)(Thr389) and IRS1(Ser636/639) were significantly increased following AMPK activation. To the best of our knowledge, this is the first report of heightened insulin responses of GSK3beta and p70S6K following AMPK activation. In addition, we found that AMPK inhibits insulin stimulation of IRS1-associated phosphatidylinositol 3-kinase activity, and that AMPK activates atypical protein kinase C and extracellular signal-regulated kinase in the heart. CONCLUSIONS/INTERPRETATIONS: Our data are indicative of differential effects of AMPK on the activation of components in the cardiac insulin-signalling pathway. These intriguing observations are critical for characterisation of the crosstalk between AMPK and insulin signalling.


Subject(s)
Heart/physiology , Insulin/physiology , Multienzyme Complexes/physiology , Myocardium/enzymology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , AMP-Activated Protein Kinases , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Blood Glucose/analysis , Enzyme Activation/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Insulin/blood , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Male , Metformin/pharmacology , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/physiology , Phosphoproteins/physiology , Phosphorylation/drug effects , Protein Kinase C/physiology , Rats , Rats, Sprague-Dawley , Ribonucleotides/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
4.
Am J Physiol Heart Circ Physiol ; 281(4): H1561-7, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11557544

ABSTRACT

We tested the hypothesis that myocardial substrate supply regulates fatty acid oxidation independent of changes in acetyl-CoA carboxylase (ACC) and 5'-AMP-activated protein kinase (AMPK) activities. Fatty acid oxidation was measured in isolated working rat hearts exposed to different concentrations of exogenous long-chain (0.4 or 1.2 mM palmitate) or medium-chain (0.6 or 2.4 mM octanoate) fatty acids. Fatty acid oxidation was increased with increasing exogenous substrate concentration in both palmitate and octanoate groups. Malonyl-CoA content only rose as acetyl-CoA supply from octanoate oxidation increased. The increases in octanoate oxidation and malonyl-CoA content were independent of changes in ACC and AMPK activity, except that ACC activity increased with very high acetyl-CoA supply levels. Our data suggest that myocardial substrate supply is the primary mechanism responsible for alterations in fatty acid oxidation rates under nonstressful conditions and when substrates are present at physiological concentrations. More extreme variations in substrate supply lead to changes in fatty acid oxidation by the additional involvement of intracellular regulatory pathways.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Fatty Acids/metabolism , Myocardium/metabolism , Acetyl Coenzyme A/metabolism , Acetyl-CoA Carboxylase/metabolism , Adenylate Kinase/metabolism , Aminoimidazole Carboxamide/pharmacology , Animals , Glycolysis , In Vitro Techniques , Male , Malonyl Coenzyme A/metabolism , Oxidation-Reduction/drug effects , Rats , Rats, Sprague-Dawley , Ribonucleotides/pharmacology , Substrate Specificity
5.
Am J Physiol Endocrinol Metab ; 279(3): E487-93, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10950814

ABSTRACT

We determined the effect of insulin on the fate of glucose and contractile function in isolated working hypertrophied hearts from rats with an aortic constriction (n = 27) and control hearts from sham-operated rats (n = 27). Insulin increased glycolysis and glycogen in control and hypertrophied hearts. The change in glycogen was brought about by increased glycogen synthesis and decreased glycogenolysis in both groups. However, the magnitude of change in glycolysis, glycogen synthesis, and glycogenolysis caused by insulin was lower in hypertrophied hearts than in control hearts. Insulin also increased glucose oxidation and contractile function in control hearts but not in hypertrophied hearts. Protein content of glucose transporters, protein kinase B, and phosphatidylinositol 3-kinase was not different between the two groups. Thus hypertrophied hearts are less responsive to the metabolic and functional effects of insulin. The reduced responsiveness involves multiple aspects of glucose metabolism, including glycolysis, glucose oxidation, and glycogen metabolism. The absence of changes in content of key regulatory molecules indicates that other sites, pathways, or factors regulating glucose utilization are responsible for these findings.


Subject(s)
Cardiomegaly/physiopathology , Insulin/physiology , Protein Serine-Threonine Kinases , Animals , Cardiomegaly/metabolism , Glucose/metabolism , Glycogen/metabolism , Glycolysis/drug effects , Glycolysis/physiology , Hemodynamics/drug effects , Hemodynamics/physiology , Immunoblotting , In Vitro Techniques , Insulin/pharmacology , Male , Monosaccharide Transport Proteins/metabolism , Organ Size , Oxidation-Reduction , Perfusion , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...