Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 151
Filter
1.
Cell Death Differ ; 22(7): 1106-16, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25361084

ABSTRACT

Glucocorticoid levels rise dramatically in late gestation to mature foetal organs in readiness for postnatal life. Immature heart function may compromise survival. Cardiomyocyte glucocorticoid receptor (GR) is required for the structural and functional maturation of the foetal heart in vivo, yet the molecular mechanisms are largely unknown. Here we asked if GR activation in foetal cardiomyocytes in vitro elicits similar maturational changes. We show that physiologically relevant glucocorticoid levels improve contractility of primary-mouse-foetal cardiomyocytes, promote Z-disc assembly and the appearance of mature myofibrils, and increase mitochondrial activity. Genes induced in vitro mimic those induced in vivo and include PGC-1α, a critical regulator of cardiac mitochondrial capacity. SiRNA-mediated abrogation of the glucocorticoid induction of PGC-1α in vitro abolished the effect of glucocorticoid on myofibril structure and mitochondrial oxygen consumption. Using RNA sequencing we identified a number of transcriptional regulators, including PGC-1α, induced as primary targets of GR in foetal cardiomyocytes. These data demonstrate that PGC-1α is a key mediator of glucocorticoid-induced maturation of foetal cardiomyocyte structure and identify other candidate transcriptional regulators that may play critical roles in the transition of the foetal to neonatal heart.


Subject(s)
Fetal Heart/physiology , Glucocorticoids/pharmacology , Mitochondria/metabolism , Myocytes, Cardiac/physiology , Transcription Factors/physiology , Animals , Gene Expression Regulation, Developmental , Mice , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Receptors, Glucocorticoid/metabolism , Signal Transduction
2.
Clin Transl Oncol ; 16(8): 746-52, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24338509

ABSTRACT

AIM: The aim of this study was to compare the effects and mechanisms of action of metformin on estrogen receptor (ER)-positive and ER-negative breast cancer cell lines. METHODS: The anti-proliferative effects of metformin, and of the direct activator of adenosine monophosphate-activated protein kinase (AMPK), A-769662, on MCF-7 (ER-positive) and MDA-MB-231 (ER-negative) breast cancer cell lines were evaluated by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide, a yellow tetrazole) assays. Fluorescence-activated cell sorting was also used to examine the effect of metformin on the cell cycle. Finally, phosphorylation of the metformin target AMPK, and of its potential downstream targets including acetyl-CoA carboxylase (ACC), p53, p70-S6K and Raptor, was examined using immunoblotting. RESULTS: Metformin and A-769662 caused significant, concentration-dependent suppression of cell proliferation with G1 cell cycle arrest in both MCF-7 and MDA-MB-231 cells. The proliferation suppression effect was more profound in MCF-7 cells. A concentration-dependent phosphorylation of AMPK was detected following metformin treatment, as was phosphorylation of ACC in both cell lines, but not p53, p70-S6k or Raptor. CONCLUSION: Metformin acts as a growth inhibitor in both ER-positive and ER-negative breast cancer cells in vitro, and arrests cells in G1 phase, particularly in the ER-positive MCF-7 cells. The effect is likely to be mediated by AMPK activation, in part by inhibition of fatty acid synthesis via ACC phosphorylation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Signal Transduction/drug effects , Blotting, Western , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Separation , Female , Flow Cytometry , Humans , Immunohistochemistry
3.
Article in English | MEDLINE | ID: mdl-22071265

ABSTRACT

Adenosine monophosphate-activated protein kinase (AMPK) is a cellular energy sensor activated by metabolic stresses that inhibit catabolic ATP production or accelerate ATP consumption. Once activated, AMPK switches on catabolic pathways, generating ATP, while inhibiting cell growth and proliferation, thus promoting energy homeostasis. AMPK is activated by the antidiabetic drug metformin, and by many natural products including "nutraceuticals" and compounds used in traditional medicines. Most of these xenobiotics activate AMPK by inhibiting mitochondrial ATP production. AMPK activation by metabolic stress requires the upstream kinase, LKB1, whose tumor suppressor effects may be largely mediated by AMPK. However, many tumor cells appear to have developed mechanisms to reduce AMPK activation and thus escape its growth-restraining effects. A similar phenomenon occurs during viral infection. If we can establish how down-regulation occurs in tumors and virus-infected cells, there may be therapeutic avenues to reverse these effects.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diabetes Mellitus/enzymology , Metabolic Networks and Pathways , Neoplasms/enzymology , Virus Diseases/enzymology , AMP-Activated Protein Kinases/chemistry , Animals , Humans , Stress, Physiological
4.
Diabetologia ; 54(9): 2392-403, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21667214

ABSTRACT

AIMS/HYPOTHESIS: Urocortins are the endogenous ligands for the corticotropin-releasing factor receptor type 2 (CRFR2), which is implicated in regulating energy balance and/or glucose metabolism. We determined the effects of chronic CRFR2 activation on metabolism in vivo, by generating and phenotyping transgenic mice overproducing the specific CRFR2 ligand urocortin 3. METHODS: Body composition, glucose metabolism, insulin sensitivity, energy efficiency and expression of key metabolic genes were assessed in adult male urocortin 3 transgenic mice (Ucn3(+)) under control conditions and following an obesogenic high-fat diet (HFD) challenge. RESULTS: Ucn3(+) mice had increased skeletal muscle mass with myocyte hypertrophy. Accelerated peripheral glucose disposal, increased respiratory exchange ratio and hypoglycaemia on fasting demonstrated increased carbohydrate metabolism. Insulin tolerance and indices of insulin-stimulated signalling were unchanged, indicating these effects were not mediated by increased insulin sensitivity. Expression of the transgene in Crfr2 (also known as Crhr2)-null mice negated key aspects of the Ucn3(+) phenotype. Ucn3(+) mice were protected from the HFD-induced hyperglycaemia and increased adiposity seen in control mice despite consuming more energy. Expression of uncoupling proteins 2 and 3 was higher in Ucn3(+) muscle, suggesting increased catabolic processes. IGF-1 abundance was upregulated in Ucn3(+) muscle, providing a potential paracrine mechanism in which urocortin 3 acts upon CRFR2 to link the altered metabolism and muscular hypertrophy observed. CONCLUSIONS/INTERPRETATION: Urocortin 3 acting on CRFR2 in skeletal muscle of Ucn3(+) mice results in a novel metabolically favourable phenotype, with lean body composition and protection against diet-induced obesity and hyperglycaemia. Urocortins and CRFR2 may be of interest as potential therapeutic targets for obesity.


Subject(s)
Dietary Fats/adverse effects , Hyperglycemia/metabolism , Hyperglycemia/prevention & control , Obesity/metabolism , Obesity/prevention & control , Urocortins/genetics , Urocortins/metabolism , Animals , Body Composition/drug effects , Body Composition/physiology , Dietary Fats/pharmacology , Disease Models, Animal , Energy Metabolism/drug effects , Energy Metabolism/physiology , Glucose/metabolism , Insulin/blood , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Phenotype , Receptors, Corticotropin-Releasing Hormone/deficiency , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism
5.
Biochim Biophys Acta ; 1804(3): 581-91, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19778642

ABSTRACT

AMP-activated protein kinase (AMPK) is a cellular energy sensor activated by metabolic stresses that either inhibit ATP synthesis or accelerate ATP consumption. Activation of AMPK in response to an increase in the cellular AMP:ATP ratio results in inhibition of ATP-consuming processes such as gluconeogenesis and fatty acid synthesis, while stimulating ATP-generating processes, including fatty acid oxidation. These alterations in lipid and glucose metabolism would be expected to ameliorate the pathogenesis of obesity, type 2 diabetes and other metabolic disorders. Recently, AMPK has also been identified as a potential target for cancer prevention and/or treatment. Cell growth and proliferation are energetically demanding, and AMPK may act as an "energy checkpoint" that permits growth and proliferation only when energy reserves are sufficient. Thus, activators of AMPK could have potential as novel therapeutics both for metabolic disorders and for cancer, which together constitute two of the most prevalent groups of diseases worldwide.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Drug Delivery Systems/methods , Enzyme Activators/therapeutic use , Metabolic Diseases/drug therapy , Neoplasms/prevention & control , Animals , Cell Proliferation/drug effects , Drug Design , Energy Metabolism/drug effects , Enzyme Activators/chemistry , Fatty Acids/biosynthesis , Fatty Acids/metabolism , Gluconeogenesis/drug effects , Humans , Metabolic Diseases/enzymology , Neoplasms/enzymology
6.
Adv Exp Med Biol ; 648: 57-63, 2009.
Article in English | MEDLINE | ID: mdl-19536465

ABSTRACT

Hypoxic inhibition of K(+) channels in type I cells is believed to be of central importance in carotid body chemotransduction. We have recently suggested that hypoxic channel inhibition is mediated by AMP-activated protein kinase (AMPK). Here, we have further explored the modulation by AMPK of recombinant K(+) channels (expressed in HEK293 cells) whose native counterparts are considered O(2)-sensitive in the rat carotid body. Inhibition of maxiK channels by AMPK activation with AICAR was found to be independent of [Ca(2+)](i) and occurred regardless of whether the alpha subunit was co-expressed with an auxiliary beta subunit. All effects of AICAR were fully reversed by the AMPK inhibitor compound C. MaxiK channels were also inhibited by the novel AMPK activator A-769662 and by intracellular dialysis with the constitutively active, truncated AMPK mutant, T172D. The molecular identity of the O(2)-sensitive leak K(+) conductance in rat type I cells remains unclear, but shares similarities with TASK-1 and TASK-3. Recombinant TASK-1 was insensitive to AICAR. However, TASK-3 was inhibited by either AICAR or A-769662 in a manner which was reversed by compound C. These data highlight a role for AMPK in the modulation of two proposed O(2) sensitive K(+) channels found in the carotid body.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Oxygen/metabolism , Potassium Channels/metabolism , AMP-Activated Protein Kinases/genetics , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Cell Line , Dialysis , Electric Conductivity , Enzyme Activation , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/metabolism , Ribonucleotides/pharmacology
7.
Diabetologia ; 52(5): 891-900, 2009 May.
Article in English | MEDLINE | ID: mdl-19252894

ABSTRACT

AIMS/HYPOTHESIS: TBC1 domain family, member 4 (TBC1D4; also known as AS160) is a cellular signalling intermediate to glucose transport regulated by insulin-dependent and -independent mechanisms. Skeletal muscle insulin sensitivity is increased after acute exercise by an unknown mechanism that does not involve modulation at proximal insulin signalling intermediates. We hypothesised that signalling through TBC1D4 is involved in this effect of exercise as it is a common signalling element for insulin and exercise. METHODS: Insulin-regulated glucose metabolism was evaluated in 12 healthy moderately trained young men 4 h after one-legged exercise at basal and during a euglycaemic-hyperinsulinaemic clamp. Vastus lateralis biopsies were taken before and immediately after the clamp. RESULTS: Insulin stimulation increased glucose uptake in both legs, with greater effects (approximately 80%, p < 0.01) in the previously exercised leg. TBC1D4 phosphorylation, assessed using the phospho-AKT (protein kinase B)substrate antibody and phospho- and site-specific antibodies targeting six phosphorylation sites on TBC1D4, increased at similar degrees to insulin stimulation in the previously exercised and rested legs (p < 0.01). However, TBC1D4 phosphorylation on Ser-318, Ser-341, Ser-588 and Ser-751 was higher in the previously exercised leg, both in the absence and in the presence of insulin (p < 0.01; Ser-588, p = 0.09; observed power = 0.39). 14-3-3 binding capacity for TBC1D4 increased equally (p < 0.01) in both legs during insulin stimulation. CONCLUSION/INTERPRETATION: We provide evidence for site-specific phosphorylation of TBC1D4 in human skeletal muscle in response to physiological hyperinsulinaemia. The data support the idea that TBC1D4 is a nexus for insulin- and exercise-responsive signals that may mediate increased insulin action after exercise.


Subject(s)
Exercise/physiology , GTPase-Activating Proteins/physiology , Insulin/physiology , Muscle, Skeletal/physiology , Adipose Tissue/cytology , Adipose Tissue/physiology , Adult , Biopsy , Blood Glucose/metabolism , DNA Primers , Diet , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Humans , Hyperinsulinism/etiology , Knee Joint/physiology , Leg/physiology , Male , Oxygen Consumption , Phosphorylation , Rest , Signal Transduction , Supine Position , Workload , Young Adult
8.
Acta Physiol (Oxf) ; 196(1): 99-113, 2009 May.
Article in English | MEDLINE | ID: mdl-19245651

ABSTRACT

The classical role of the AMP-activated protein kinase (AMPK) is to act as a sensor of the immediate availability of cellular energy, by monitoring the concentrations of AMP and ATP. However, the beta subunits of AMPK contain a glycogen-binding domain, and in this review we develop the hypothesis that this is a regulatory domain that allows AMPK to act as a sensor of the status of cellular reserves of energy in the form of glycogen. We argue that the pool of AMPK that is bound to the glycogen particle is in an active state when glycogen particles are fully synthesized, causing phosphorylation of glycogen synthase at site 2 and providing a feedback inhibition of further extension of the outer chains of glycogen. However, when glycogen becomes depleted, the glycogen-bound pool of AMPK becomes inhibited due to binding to alpha1-->6-linked branch points exposed by the action of phosphorylase and/or debranching enzyme. This allows dephosphorylation of site 2 on glycogen synthase by the glycogen-bound form of protein phosphatase-1, promoting rapid resynthesis of glycogen and replenishment of glycogen stores. This is an extension of the classical role of AMPK as a 'guardian of cellular energy', in which it ensures that cellular energy reserves are adequate for medium-term requirements. The literature concerning AMPK, glycogen structure and glycogen-binding proteins that led us to this concept is reviewed.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Glycogen/metabolism , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/genetics , Animals , Binding Sites , Glycogen/chemistry , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Humans , Protein Binding , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism
9.
Int J Obes (Lond) ; 32 Suppl 4: S7-12, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18719601

ABSTRACT

The AMP-activated protein kinase (AMPK) system is a key player in regulating energy balance at both the cellular and whole-body levels, placing it at centre stage in studies of obesity, diabetes and the metabolic syndrome. It is switched on in response to metabolic stresses such as muscle contraction or hypoxia, and modulated by hormones and cytokines affecting whole-body energy balance such as leptin, adiponectin, resistin, ghrelin and cannabinoids. Once activated, it switches on catabolic pathways that generate adenosine triphosphate (ATP), while switching off ATP-consuming anabolic processes. AMPK exists as heterotrimeric complexes comprising a catalytic alpha-subunit and regulatory beta- and gamma-subunits. Binding of AMP to the gamma-subunit, which is antagonized by high ATP, causes activation of the kinase by promoting phosphorylation at threonine (Thr-172) on the alpha-subunit by the upstream kinase LKB1, allowing the system to act as a sensor of cellular energy status. In certain cells, AMPK is activated in response to elevation of cytosolic Ca2+ via phosphorylation of Thr-172 by calmodulin-dependent kinase kinase-beta (CaMKKbeta). Activation of AMPK, either in response to exercise or to pharmacological agents, has considerable potential to reverse the metabolic abnormalities associated with type 2 diabetes and the metabolic syndrome. Two existing classes of antidiabetic drugs, that is, biguanides (for example, metformin) and the thiazolidinediones (for example, rosiglitazone), both act (at least in part) by activation of AMPK. Novel drugs activating AMPK may also have potential for the treatment of obesity.


Subject(s)
AMP-Activated Protein Kinases/physiology , Diabetes Mellitus/enzymology , Energy Metabolism/physiology , Metabolic Syndrome/enzymology , Protein Serine-Threonine Kinases/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Kinase , Glucose/metabolism , Humans , Mice , Signal Transduction/physiology , Stress, Physiological/physiology
10.
Br J Pharmacol ; 153(8): 1669-77, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18297106

ABSTRACT

BACKGROUND AND PURPOSE: Galegine and guanidine, originally isolated from Galega officinalis, led to the development of the biguanides. The weight-reducing effects of galegine have not previously been studied and the present investigation was undertaken to determine its mechanism(s) of action. EXPERIMENTAL APPROACH: Body weight and food intake were examined in mice. Glucose uptake and acetyl-CoA carboxylase activity were studied in 3T3-L1 adipocytes and L6 myotubes and AMP activated protein kinase (AMPK) activity was examined in cell lines. The gene expression of some enzymes involved in fat metabolism was examined in 3T3-L1 adipocytes. KEY RESULTS: Galegine administered in the diet reduced body weight in mice. Pair-feeding indicated that at least part of this effect was independent of reduced food intake. In 3T3-L1 adipocytes and L6 myotubes, galegine (50 microM-3 mM) stimulated glucose uptake. Galegine (1-300 microM) also reduced isoprenaline-mediated lipolysis in 3T3-L1 adipocytes and inhibited acetyl-CoA carboxylase activity in 3T3-L1 adipocytes and L6 myotubes. Galegine (500 microM) down-regulated genes concerned with fatty acid synthesis, including fatty acid synthase and its upstream regulator SREBP. Galegine (10 microM and above) produced a concentration-dependent activation of AMP activated protein kinase (AMPK) in H4IIE rat hepatoma, HEK293 human kidney cells, 3T3-L1 adipocytes and L6 myotubes. CONCLUSIONS AND IMPLICATIONS: Activation of AMPK can explain many of the effects of galegine, including enhanced glucose uptake and inhibition of acetyl-CoA carboxylase. Inhibition of acetyl-CoA carboxylase both inhibits fatty acid synthesis and stimulates fatty acid oxidation, and this may to contribute to the in vivo effect of galegine on body weight.


Subject(s)
Eating/drug effects , Guanidines/pharmacology , Multienzyme Complexes/drug effects , Protein Serine-Threonine Kinases/drug effects , Weight Loss/drug effects , AMP-Activated Protein Kinases , Acetyl-CoA Carboxylase/antagonists & inhibitors , Acetyl-CoA Carboxylase/metabolism , Animals , Cell Line , Fatty Acids/metabolism , Galega/chemistry , Gene Expression Regulation, Enzymologic/drug effects , Glucose/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats
11.
Br J Pharmacol ; 151(8): 1204-15, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17603555

ABSTRACT

BACKGROUND AND PURPOSE: AMP-activated protein kinase (AMPK) is activated by metformin, phenformin, and the AMP mimetic, 5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside (AICAR). We have completed an extensive study of the pharmacological effects of these drugs on AMPK activation, adenine nucleotide concentration, transepithelial amiloride-sensitive (I(amiloride)) and ouabain-sensitive basolateral (I(ouabain)) short circuit current in H441 lung epithelial cells. EXPERIMENTAL APPROACH: H441 cells were grown on permeable filters at air interface. I(amiloride), I(ouabain) and transepithelial resistance were measured in Ussing chambers. AMPK activity was measured as the amount of radiolabelled phosphate transferred to the SAMS peptide. Adenine nucleotide concentration was analysed by reverse phase HPLC and NAD(P)H autofluorescence was measured using confocal microscopy. KEY RESULTS: Phenformin, AICAR and metformin increased AMPK (alpha1) activity and decreased I(amiloride). The AMPK inhibitor Compound C prevented the action of metformin and AICAR but not phenformin. Phenformin and AICAR decreased I(ouabain) across H441 monolayers and decreased monolayer resistance. The decrease in I(amiloride) was closely related to I(ouabain) with phenformin, but not in AICAR treated monolayers. Metformin and phenformin increased the cellular AMP:ATP ratio but only phenformin and AICAR decreased cellular ATP. CONCLUSIONS AND IMPLICATIONS: Activation of alpha1-AMPK is associated with inhibition of apical amiloride-sensitive Na(+) channels (ENaC), which has important implications for the clinical use of metformin. Additional pharmacological effects evoked by AICAR and phenformin on I(ouabain), with potential secondary effects on apical Na+ conductance, ENaC activity and monolayer resistance, have important consequences for their use as pharmacological activators of AMPK in cell systems where Na+K+ATPase is an important component.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Multienzyme Complexes/drug effects , Phenformin/pharmacology , Protein Serine-Threonine Kinases/drug effects , Ribonucleotides/pharmacology , Sodium/metabolism , AMP-Activated Protein Kinases , Adenine Nucleotides/metabolism , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Amiloride , Aminoimidazole Carboxamide/pharmacology , Cell Line , Chromatography, High Pressure Liquid , Epithelial Cells , Epithelial Sodium Channels/drug effects , Fluorescence , Humans , Lung , Microscopy, Confocal , Multienzyme Complexes/metabolism , Ouabain , Protein Serine-Threonine Kinases/metabolism
13.
Int J Obes Relat Metab Disord ; 28(6): 824-8, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15024403

ABSTRACT

OBJECTIVE: An unexplained phenotype of mice overexpressing human UCP3 is their improved glucose homeostasis. Since overexpression of UCP3 might affect the energy charge of the cell, we investigated whether these mice have an increased AMP-activated protein kinase (AMPK) activity. METHODS: Mitochondrial localisation of UCP3 was determined by immunoelectronmicroscopy and AMPK activity was measured in medial gastrocnemius of control mice and mice overexpressing human UCP3. RESULTS: Mice overexpressing human UCP3 had 5.8 fold higher levels of UCP3 protein, for which mitochondrial localisation was confirmed by immunoelectronmicroscopy. The ATP/AMP ratio was significantly lower in mice over-expressing UCP3 compared to the wild-type (10.9+/-1.6 vs 20.4+/-1.9 AU, P=0.03). Over-expression of UCP3 resulted in increased AMPK alpha1 activity (1.23+/-0.05 vs 1.00+/-0.06 normalized values, P=0.004) and a tendency towards increased AMPK alpha2 activity (1.18+/-0.08 vs 1.00+/-0.10 normalized values, P=0.08). CONCLUSION: Increased AMPK activity provides a plausible explanation for the improved glucose tolerance characteristic for these mice.


Subject(s)
Adenylate Kinase/metabolism , Carrier Proteins/analysis , Glucose/metabolism , Homeostasis/physiology , Adenine Nucleotides/metabolism , Animals , Carrier Proteins/genetics , Energy Metabolism , Ion Channels , Mice , Mice, Inbred Strains , Microscopy, Immunoelectron/methods , Mitochondrial Proteins , Phenotype , Uncoupling Agents/analysis , Uncoupling Protein 3
14.
Biochem Soc Trans ; 31(Pt 1): 182-5, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12546681

ABSTRACT

Evidence is accumulating for roles of AMP-activated protein kinase (AMPK) in controlling glucose uptake, fatty acid oxidation and gene expression in skeletal muscle. Relatively little is known, however, about the control of expression of the AMPK subunit isoforms. Marked differences are noted in subunit expression as a function of muscle fibre type. Expression of the gamma3 subunit isoform increases in fast-twitch red fibres of the rat in response to training. All subunit isoforms are expressed to a lesser extent in rats treated with propylthiouracil (PTU; an inhibitor of thyroid hormone synthesis) for 3 weeks compared with rats given excess thyroid hormones for 3 weeks. An approx. 2-fold increase in acetyl-CoA carboxylase was observed in gastrocnemius of hyperthyroid rats compared with experimentally hypothyroid rats. Thyroid state therefore appears to be one important factor controlling expression of these proteins in skeletal muscle.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Gene Expression Regulation, Enzymologic , Multienzyme Complexes/metabolism , Muscle, Skeletal/enzymology , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Animals , Blotting, Western , Liver/metabolism , Muscle, Skeletal/metabolism , Protein Isoforms , Rats , Thyroid Hormones/metabolism , Time Factors
15.
J Appl Physiol (1985) ; 93(6): 2081-8, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12433937

ABSTRACT

AMP-activated protein kinase (AMPK) consists of three subunits: alpha, beta, and gamma. Two isoforms exist for the alpha-subunit (alpha(1) and alpha(2)), two for the beta-subunit (beta(1) and beta(2)), and three for the gamma-subunit (gamma(1), gamma(2), and gamma(3)). Although the specific roles of the beta- and gamma-subunits are not well understood, the alpha-subunit isoforms contain the catalytic site and also the phosphorylation/activation site for the upstream kinase. This study was designed to determine the role of thyroid hormones in controlling expression levels of these AMPK subunits and of one downstream target, acetyl-CoA carboxylase (ACC), in muscle. AMPK subunit and ACC levels were determined by Western blots in control rats, in rats given 0.01% propylthiouracil (PTU) in drinking water for 3 wk, and in rats given 3 mg of thyroxine and 1 mg of triiodothyronine per kilogram chow for 1 or 3 wk. In gastrocnemius muscle, all isoforms of AMPK subunits were significantly increased in rats given thyroid hormones for 3 wk vs. those treated with PTU. Similar patterns were seen in individual muscle types. Expression of muscle ACC was also significantly increased in response to 3 wk of treatment with excess thyroid hormones. Muscle content of malonyl-CoA was elevated in PTU-treated rats and depressed in thyroid hormone-treated rats. These data provide evidence that skeletal muscle AMPK subunit and ACC expression is partially under the control of thyroid hormones.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Multienzyme Complexes/metabolism , Muscle, Skeletal/enzymology , Protein Serine-Threonine Kinases/metabolism , Thyroid Gland/physiology , AMP-Activated Protein Kinases , Adipose Tissue/physiology , Animals , Antithyroid Agents/pharmacology , Blotting, Western , Body Weight/drug effects , Body Weight/physiology , Eating/drug effects , Eating/physiology , Glycogen/metabolism , Male , Malonyl Coenzyme A/metabolism , Muscle, Skeletal/drug effects , Phosphorylation , Propylthiouracil/pharmacology , Rats , Rats, Sprague-Dawley , Thyroxine/pharmacology , Triiodothyronine/pharmacology
16.
Biochem Soc Trans ; 30(Pt 6): 1064-70, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12440973

ABSTRACT

The AMP-activated protein kinase (AMPK) is a sensor of cellular energy charge and a 'metabolic master switch'. When activated by ATP depletion, it switches off ATP-consuming processes, while switching on catabolic pathways that generate ATP. AMPK exists as heterotrimeric complexes comprising catalytic alpha subunits and regulatory beta and gamma subunits, each of which occurs as multiple isoforms. Rising AMP and falling ATP, brought about by various types of cellular stress (including exercise in skeletal muscle), stimulate the system in an ultrasensitive manner. Acetyl-CoA carboxylase (ACC) exists in mammals as two isoforms, termed ACC-1 and ACC-2 (also known as ACC-alpha and ACC-beta). AMPK phosphorylates and inactivates both isoforms at the equivalent site. Knockout mice, and other approaches, suggest that the malonyl-CoA produced by ACC-2 is exclusively involved in regulation of fatty acid oxidation, whereas that produced by ACC-1 is utilized in fatty acid synthesis. Activation of AMPK by cellular stress or exercise therefore switches on fatty acid oxidation (via phosphorylation of ACC-2) while switching off fatty acid synthesis (via phosphorylation of ACC-1). The Drosophila melanogaster genome contains single genes encoding homologues of the alpha, beta and gamma subunits of AMPK (DmAMPK) and of ACC (DmACC). Studies in a Drosophila embryonal cell line show that DmAMPK is activated by stresses that cause ATP depletion (oligomycin, hypoxia or glucose deprivation) and that this is associated with phosphorylation of the site on DmACC equivalent to the AMPK sites on mammalian ACC-1 and ACC-2. This is abolished when expression of DmAMPK is ablated using an RNA interference approach, proving that DmAMPK is necessary for phosphorylation of DmACC in response to ATP depletion.


Subject(s)
Fatty Acids/metabolism , Multienzyme Complexes/metabolism , Oxygen/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Amino Acid Sequence , Animals , Drosophila melanogaster/metabolism , Humans , Models, Genetic , Molecular Sequence Data , Peptides , Phosphorylation , Protein Isoforms , Protein Structure, Tertiary , Rats , Sequence Homology, Amino Acid
17.
Bioessays ; 23(12): 1112-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11746230

ABSTRACT

The AMP-activated protein kinase cascade is a sensor of cellular energy charge, and its existence provides strong support for the energy charge hypothesis first proposed by Daniel Atkinson in the 1960s. The system is activated in an ultrasensitive manner by cellular stresses that deplete ATP (and consequently elevate AMP), either by inhibiting ATP production (e.g., hypoxia), or by accelerating ATP consumption (e.g., exercise in muscle). Once activated, it switches on catabolic pathways, both acutely by phosphorylation of metabolic enzymes and chronically by effects on gene expression, and switches off many ATP-consuming processes. Recent work suggests that activation of AMPK is responsible for many of the effects of physical exercise, both the rapid metabolic effects and the adaptations that occur during training. Dominant mutations in regulatory subunit isoforms (gamma2 and gamma3) of AMPK, which appear to increase the basal activity in the absence of AMP, lead to hypertrophy of cardiac and skeletal muscle respectively.


Subject(s)
Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Amino Acid Sequence , Animals , Energy Metabolism , Humans , Metabolic Diseases/enzymology , Models, Molecular , Molecular Sequence Data , Multienzyme Complexes/chemistry , Multienzyme Complexes/physiology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/physiology
18.
J Biol Chem ; 276(50): 46912-6, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11598104

ABSTRACT

Exercise is known to increase insulin sensitivity and is an effective form of treatment for the hyperglycemia observed in type 2 diabetes. Activation of 5'-AMP-activated protein kinase (AMPK) by 5-aminoimidazole-4-carboxamide riboside (AICAR), exercise, or electrically stimulated contraction leads to increased glucose transport in skeletal muscle. Here we report the first evidence of a direct interaction between AMPK and the most upstream component of the insulin-signaling cascade, insulin receptor substrate-1 (IRS-1). We find that AMPK rapidly phosphorylates IRS-1 on Ser-789 in cell-free assays as well as in mouse C2C12 myotubes incubated with AICAR. In the C2C12 myotubes activation of AMPK by AICAR matched the phosphorylation of IRS-1 on Ser-789. This phosphorylation correlates with a 65% increase in insulin-stimulated IRS-1-associated phosphatidylinositol 3-kinase activity in C2C12 myotubes preincubated with AICAR. The binding of phosphatidylinositol 3-kinase to IRS-1 was not affected by AICAR. These results demonstrate the existence of an interaction between AMPK and early insulin signaling that could be of importance to our understanding of the potentiating effects of exercise on insulin signaling.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/chemistry , Multienzyme Complexes/metabolism , Myocardium/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribonucleosides/chemistry , Serine/chemistry , AMP-Activated Protein Kinases , Animals , Binding Sites , Biological Transport , Blotting, Western , Cells, Cultured , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Glucose/metabolism , Humans , Insulin/metabolism , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Mice , Muscle, Skeletal , Myocardium/cytology , Peptides/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Binding , Recombinant Proteins/metabolism , Signal Transduction , Subcellular Fractions , Time Factors
19.
FEBS Lett ; 505(3): 348-52, 2001 Sep 21.
Article in English | MEDLINE | ID: mdl-11576526

ABSTRACT

AMP-activated protein kinase (AMPK) is known to be activated by phosphorylation on Thr172 in response to an increased AMP/ATP ratio. We report here that such an activation indeed occurred in anaerobic rat hearts and that it was antagonized (40-50%) when the hearts were pre-treated with 100 nM insulin. The effect of insulin (1) was blocked by wortmannin, an inhibitor of phosphatidylinositol-3-kinase; (2) only occurred when insulin was added before anoxia, suggesting a hierarchical control; (3) resulted in a decreased phosphorylation state of Thr172 in AMPK and (4) was unrelated to changes in the AMP/ATP ratio. This is the first demonstration that AMPK activity could be changed without a detectable change in the AMP/ATP ratio of the cardiac cell.


Subject(s)
Cell Hypoxia , Insulin/pharmacology , Multienzyme Complexes/antagonists & inhibitors , Myocardial Ischemia/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Enzyme Activation , Enzyme Inhibitors/pharmacology , Male , Multienzyme Complexes/metabolism , Myocardium/cytology , Myocardium/enzymology , Myocardium/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Wistar
20.
Plant Mol Biol ; 45(6): 731-41, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11430435

ABSTRACT

Using a PCR approach, we have cloned DNA encoding a catalytic subunit isoform (SnRK1-alpha1) of SNF1-related protein kinase-1 from spinach leaf. The predicted amino acid sequence falls into the SnRK1a sub-family, and is closely related to SnRK1a sequences expressed in cucumber, Arabidopsis thaliana, tobacco and potato. We have generated two affinity-purified antipeptide antibodies (anti-RASS and anti-AEF) based on the predicted amino acid sequence of spinach SnRK1-alpha1. They were used to analyse multiple forms of SNF1-related kinase (HRK-A, -C, -D) that were previously identified by biochemical criteria in extracts of spinach leaf (Sugden et al., Plant Physiol. 120 (1999), 257-274). Anti-AEF appears to be specific for the SnRK1-alpha1 isoform, whereas anti-RASS is a 'pan-alpha' antibody that precipitates all isoforms present in spinach leaf extracts. The activities of HRK-A and HRK-C can be entirely accounted for by the SnRK1-alpha1 catalytic subunit. By contrast, only a small proportion of HRK-D activity (ca. 20%) can be accounted for by SnRK1-alpha1, with the remainder presumably being due to other isoforms (SnRK1-alpha2?) that are currently poorly defined. A 35 kDa polypeptide recognized by an antibody against the putative Arabidopsis beta2 subunit co-precipitates with HRK-C, but not HRK-A or D.


Subject(s)
Protein Serine-Threonine Kinases/genetics , Spinacia oleracea/enzymology , Amino Acid Sequence , Base Sequence , Blotting, Western , Catalytic Domain , Cloning, Molecular , DNA, Plant , Isoenzymes/genetics , Molecular Sequence Data , Peptides/immunology , Plant Leaves/enzymology , Precipitin Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...