Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Neurochem ; 127(1): 66-77, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23841933

ABSTRACT

Growth factors and nutrients, such as amino acids and glucose, regulate mammalian target of rapamycin complex 1 (mTORC1) signaling and subsequent translational control in a coordinated manner. Brain-derived neurotrophic factor (BDNF), the most prominent neurotrophic factor in the brain, activates mTORC1 and induces phosphorylation of its target, p70S6 kinase (p70S6K), at Thr389 in neurons. BDNF also increases mammalian target of rapamycin-dependent novel protein synthesis in neurons. Here, we report that BDNF-induced p70S6K activation is dependent on glucose, but not amino acids, sufficiency in cultured cortical neurons. AMP-activated protein kinase (AMPK) is the molecular background to this specific nutrient dependency. Activation of AMPK, which is induced by glucose deprivation, treatment with pharmacological agents such as 2-deoxy-D-glucose, metformin, and 5-aminoimidazole-4-carboxamide ribonucleoside or forced expression of a constitutively active AMPKα subunit, counteracts BDNF-induced phosphorylation of p70S6K and enhanced protein synthesis in cortical neurons. These results indicate that AMPK inhibits the effects of BDNF on mTORC1-mediated translation in neurons.


Subject(s)
AMP-Activated Protein Kinases/physiology , Brain-Derived Neurotrophic Factor/pharmacology , Multiprotein Complexes/physiology , Neurons/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/physiology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Deoxyglucose/pharmacology , Electrophoresis, Polyacrylamide Gel , Electroporation , Fibroblasts/metabolism , Glucose/deficiency , Glucose/physiology , Hypoglycemic Agents/pharmacology , Immunohistochemistry , Immunoprecipitation , Mechanistic Target of Rapamycin Complex 1 , Metformin/pharmacology , Methionine/metabolism , Oncogene Protein v-akt/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
2.
Mol Cell Biol ; 29(13): 3455-64, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19380482

ABSTRACT

Hypoxia promotes Na,K-ATPase endocytosis via protein kinase C zeta (PKC zeta)-mediated phosphorylation of the Na,K-ATPase alpha subunit. Here, we report that hypoxia leads to the phosphorylation of 5'-AMP-activated protein kinase (AMPK) at Thr172 in rat alveolar epithelial cells. The overexpression of a dominant-negative AMPK alpha subunit (AMPK-DN) construct prevented the hypoxia-induced endocytosis of Na,K-ATPase. The overexpression of the reactive oxygen species (ROS) scavenger catalase prevented hypoxia-induced AMPK activation. Moreover, hypoxia failed to activate AMPK in mitochondrion-deficient rho(0)-A549 cells, suggesting that mitochondrial ROS play an essential role in hypoxia-induced AMPK activation. Hypoxia-induced PKC zeta translocation to the plasma membrane and phosphorylation at Thr410 were prevented by the pharmacological inhibition of AMPK or by the overexpression of the AMPK-DN construct. We found that AMPK alpha phosphorylates PKC zeta on residue Thr410 within the PKC zeta activation loop. Importantly, the activation of AMPK alpha was necessary for hypoxia-induced AMPK-PKC zeta binding in alveolar epithelial cells. The overexpression of T410A mutant PKC zeta prevented hypoxia-induced Na,K-ATPase endocytosis, confirming that PKC zeta Thr410 phosphorylation is essential for this process. PKC zeta activation by AMPK is isoform specific, as small interfering RNA targeting the alpha1 but not the alpha2 catalytic subunit prevented PKC zeta activation. Accordingly, we provide the first evidence that hypoxia-generated mitochondrial ROS lead to the activation of the AMPK alpha1 isoform, which binds and directly phosphorylates PKC zeta at Thr410, thereby promoting Na,K-ATPase endocytosis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Endocytosis/physiology , Epithelial Cells/metabolism , Hypoxia/metabolism , Protein Kinase C/metabolism , Pulmonary Alveoli/cytology , Sodium-Potassium-Exchanging ATPase/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Enzyme Activation , Epithelial Cells/cytology , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mitochondria/metabolism , Phosphorylation , Protein Kinase C/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Sodium-Potassium-Exchanging ATPase/genetics
3.
Cell Metab ; 7(5): 377-88, 2008 May.
Article in English | MEDLINE | ID: mdl-18460329

ABSTRACT

Detailed knowledge of the pathways by which ghrelin and leptin signal to AMPK in hypothalamic neurons and lead to regulation of appetite and glucose homeostasis is central to the development of effective means to combat obesity. Here we identify CaMKK2 as a component of one of these pathways, show that it regulates hypothalamic production of the orexigenic hormone NPY, provide evidence that it functions as an AMPKalpha kinase in the hypothalamus, and demonstrate that it forms a unique signaling complex with AMPKalpha and beta. Acute pharmacologic inhibition of CaMKK2 in wild-type mice, but not CaMKK2 null mice, inhibits appetite and promotes weight loss consistent with decreased NPY and AgRP mRNAs. Moreover, the loss of CaMKK2 protects mice from high-fat diet-induced obesity, insulin resistance, and glucose intolerance. These data underscore the potential of targeting CaMKK2 as a therapeutic intervention.


Subject(s)
Appetite Regulation/physiology , Calcium-Calmodulin-Dependent Protein Kinase Kinase/physiology , Energy Metabolism/physiology , Hypothalamus/enzymology , Insulin Resistance/physiology , AMP-Activated Protein Kinase Kinases , Acetyl-CoA Carboxylase/metabolism , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Benzimidazoles/pharmacology , Cells, Cultured , Diet, Atherogenic , Female , Glucose Intolerance/etiology , Glucose Tolerance Test , Hypothalamus/pathology , Immunoblotting , Immunoenzyme Techniques , Immunoprecipitation , In Situ Hybridization , Insulin/metabolism , Integrases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Naphthalimides/pharmacology , Neuropeptide Y/metabolism , Protein Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection , Weight Loss
4.
J Clin Invest ; 118(2): 752-62, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18188452

ABSTRACT

Hypercapnia (elevated CO(2) levels) occurs as a consequence of poor alveolar ventilation and impairs alveolar fluid reabsorption (AFR) by promoting Na,K-ATPase endocytosis. We studied the mechanisms regulating CO(2)-induced Na,K-ATPase endocytosis in alveolar epithelial cells (AECs) and alveolar epithelial dysfunction in rats. Elevated CO(2) levels caused a rapid activation of AMP-activated protein kinase (AMPK) in AECs, a key regulator of metabolic homeostasis. Activation of AMPK was mediated by a CO(2)-triggered increase in intracellular Ca(2+) concentration and Ca(2+)/calmodulin-dependent kinase kinase-beta (CaMKK-beta). Chelating intracellular Ca(2+) or abrogating CaMKK-beta function by gene silencing or chemical inhibition prevented the CO(2)-induced AMPK activation in AECs. Activation of AMPK or overexpression of constitutively active AMPK was sufficient to activate PKC-zeta and promote Na,K-ATPase endocytosis. Inhibition or downregulation of AMPK via adenoviral delivery of dominant-negative AMPK-alpha(1) prevented CO(2)-induced Na,K-ATPase endocytosis. The hypercapnia effects were independent of intracellular ROS. Exposure of rats to hypercapnia for up to 7 days caused a sustained decrease in AFR. Pretreatment with a beta-adrenergic agonist, isoproterenol, or a cAMP analog ameliorated the hypercapnia-induced impairment of AFR. Accordingly, we provide evidence that elevated CO(2) levels are sensed by AECs and that AMPK mediates CO(2)-induced Na,K-ATPase endocytosis and alveolar epithelial dysfunction, which can be prevented with beta-adrenergic agonists and cAMP.


Subject(s)
Carbon Dioxide/metabolism , Endocytosis , Hypercapnia/enzymology , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , AMP-Activated Protein Kinases , Adrenergic beta-Agonists/pharmacology , Animals , Calcium/antagonists & inhibitors , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Chelating Agents/pharmacology , Cyclic AMP/pharmacology , Endocytosis/drug effects , Endocytosis/genetics , Extracellular Fluid/metabolism , Humans , Isoproterenol/pharmacology , Protein Kinase C/metabolism , Pulmonary Alveoli/enzymology , Rats , Rats, Sprague-Dawley , Respiratory Mucosa/enzymology
5.
J Biol Chem ; 283(8): 4799-807, 2008 Feb 22.
Article in English | MEDLINE | ID: mdl-18079111

ABSTRACT

AMP-activated protein kinase (AMPK) plays multiple roles in the body's overall metabolic balance and response to exercise, nutritional stress, hormonal stimulation, and the glucose-lowering drugs metformin and rosiglitazone. AMPK consists of a catalytic alpha subunit and two non-catalytic subunits, beta and gamma, each with multiple isoforms that form active 1:1:1 heterotrimers. Here we show that recombinant human AMPK alpha1beta1gamma1 expressed in insect cells is monomeric and displays specific activity and AMP responsiveness similar to rat liver AMPK. The previously determined crystal structure of the core of mammalian alphabetagamma complex shows that beta binds alpha and gamma. Here we show that a beta1(186-270)gamma1 complex can form in the absence of detectable alpha subunit. Moreover, using alanine mutagenesis we show that beta1 Thr-263 and Tyr-267 are required for betagamma association but not alphabeta association.


Subject(s)
Liver/enzymology , Multienzyme Complexes/chemistry , Protein Serine-Threonine Kinases/chemistry , AMP-Activated Protein Kinases , Animals , COS Cells , Catalytic Domain/genetics , Chlorocebus aethiops , Exercise/physiology , Glucose/metabolism , Hormones/pharmacology , Humans , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Mutagenesis, Site-Directed , Protein Binding/drug effects , Protein Binding/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Quaternary , Rats , Rosiglitazone , Stress, Physiological/enzymology , Thiazolidinediones/pharmacology
6.
Am J Physiol Heart Circ Physiol ; 293(6): H3456-64, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17906100

ABSTRACT

Although mutations in the gamma-subunit of AMP-activated protein kinase (AMPK) can result in excessive glycogen accumulation and cardiac hypertrophy, the mechanisms by which this occurs have not been well defined. Because >65% of cardiac AMPK activity is associated with the gamma1-subunit of AMPK, we investigated the effects of expression of an AMPK-activating gamma1-subunit mutant (gamma1 R70Q) on regulatory pathways controlling glycogen accumulation and cardiac hypertrophy in neonatal rat cardiac myocytes. Whereas expression of gamma1 R70Q displayed the expected increase in palmitate oxidation rates, rates of glycolysis were significantly depressed. In addition, glycogen synthase activity was increased in cardiac myocytes expressing gamma1 R70Q, due to both increased expression and decreased phosphorylation of glycogen synthase. The inhibition of glycolysis and increased glycogen synthase activity were correlated with elevated glycogen levels in gamma1 R70Q-expressing myocytes. In association with the reduced phosphorylation of glycogen synthase, glycogen synthase kinase (GSK)-3beta protein and mRNA levels were profoundly decreased in the gamma1 R70Q-expressing myocytes. Consistent with GSK-3beta negatively regulating hypertrophy via inhibition of nuclear factor of activated T cells (NFAT), the dramatic downregulation of GSK-3beta was associated with increased nuclear activity of NFAT. Together, these data provide important new information about the mechanisms by which a mutation in the gamma-subunit of AMPK causes altered AMPK signaling and identify multiple pathways involved in regulating both cardiac myocyte metabolism and growth that may contribute to the development of the gamma mutant-associated cardiomyopathy.


Subject(s)
Glycogen/metabolism , Multienzyme Complexes/metabolism , Mutation , Myocytes, Cardiac/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , AMP-Activated Protein Kinases , Active Transport, Cell Nucleus , Animals , Animals, Newborn , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cell Size , Cells, Cultured , Glucose/metabolism , Glycogen Synthase/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Glycolysis , Hypertrophy , Multienzyme Complexes/genetics , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , NFATC Transcription Factors/metabolism , Oxidation-Reduction , Palmitic Acid/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism , Rats , Signal Transduction/genetics , Transduction, Genetic
7.
Biochem Biophys Res Commun ; 362(2): 354-9, 2007 Oct 19.
Article in English | MEDLINE | ID: mdl-17709097

ABSTRACT

AMP-activated protein kinase (AMPK) is widely recognized as an important regulator of glucose transport in skeletal muscle. The p38 mitogen-activated protein kinase (MAPK) has been proposed to be a component of AMPK-mediated signaling. Here we used several different models of altered AMPK activity to determine whether p38 MAPK is a downstream intermediate of AMPK-mediated signaling in skeletal muscle. First, L6 myoblasts and myotubes were treated with AICAR, an AMPK stimulator. AMPK phosphorylation was significantly increased, but there was no change in p38 MAPK phosphorylation. Similarly, AICAR incubation of isolated rat extensor digitorum longus (EDL) muscles did not increase p38 phosphorylation. Next, we used transgenic mice expressing an inactive form of the AMPKalpha2 catalytic subunit in skeletal muscle (AMPKalpha2i TG mice). AMPKalpha2i TG mice did not exhibit any defect in basal or contraction-induced p38 MAPK phosphorylation. We also used transgenic mice expressing an activating mutation in the AMPKgamma1 subunit (gamma1R70Q TG mice). Despite activated AMPK, basal p38 MAPK phosphorylation was not different between wild type and gamma1R70Q TG mice. In addition, muscle contraction-induced p38 MAPK phosphorylation was significantly blunted in the gamma1R70Q TG mice. In conclusion, increasing AMPK activity by AICAR and AMPKgamma1 mutation does not increase p38 MAPK phosphorylation in skeletal muscle. Furthermore, AMPKalpha2i TG mice lacking contraction-stimulated AMPK activity have normal p38 MAPK phosphorylation. These results suggest that p38 MAPK is not a downstream component of AMPK-mediated signaling in skeletal muscle.


Subject(s)
Multienzyme Complexes/metabolism , Muscle, Skeletal/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases , Amino Acid Sequence , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Cell Line , Enzyme Activation/drug effects , Immunoblotting , Mice , Mice, Inbred ICR , Mice, Transgenic , Molecular Sequence Data , Multienzyme Complexes/genetics , Muscle Contraction/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Mutation , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Rats , Rats, Sprague-Dawley , Ribonucleotides/pharmacology , p38 Mitogen-Activated Protein Kinases/genetics
8.
Diabetes ; 56(8): 2062-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17513699

ABSTRACT

Regular endurance exercise has profound benefits on overall health, including the prevention of obesity, cardiovascular disease, and diabetes. The objective of this study was to determine whether AMP-activated protein kinase (AMPK) mediates commonly observed adaptive responses to exercise training in skeletal muscle. Six weeks of voluntary wheel running induced a significant (P < 0.05) fiber type IIb to IIa/x shift in triceps muscle of wild-type mice. Despite similar wheel running capacities, this training-induced shift was reduced by approximately 40% in transgenic mice expressing a muscle-specific AMPKalpha2 inactive subunit. Sedentary mice carrying an AMPK-activating mutation (gamma1TG) showed a 2.6-fold increase in type IIa/x fibers but no further increase with training. To determine whether AMPK is involved in concomitant metabolic adaptations to training, we measured markers of mitochondria (citrate synthase and succinate dehydrogenase) and glucose uptake capacity (GLUT4 and hexokinase II). Mitochondrial markers increased similarly in wild-type and AMPKalpha2-inactive mice. Sedentary gamma1TG mice showed a approximately 25% increase in citrate synthase activity but no further increase with training. GLUT4 protein expression was not different in either line of transgenic mice compared with wild-type mice and tended to increase with training, although this increase was not statistically significant. Training induced a approximately 65% increase in hexokinase II protein in wild-type mice but not in AMPKalpha2-inactive mice. Hexokinase II was significantly elevated in sedentary gamma1TG mice, without an additional increase with training. AMPK is not necessary for exercise training-induced increases in mitochondrial markers, but it is essential for fiber type IIb to IIa/x transformation and increases in hexokinase II protein.


Subject(s)
Adaptation, Biological , Multienzyme Complexes/metabolism , Muscle, Skeletal/enzymology , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Animals , Biomarkers/metabolism , Gene Expression Regulation, Enzymologic , Glucose Transporter Type 4/metabolism , Hexokinase/metabolism , Mice , Mice, Transgenic , Mitochondrial Proteins/metabolism , Multienzyme Complexes/genetics , Mutation/genetics , Myosin Heavy Chains/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Physical Conditioning, Animal , Protein Serine-Threonine Kinases/genetics , Protein Subunits/genetics , Protein Subunits/metabolism , Trans-Activators/metabolism , Transcription Factors
9.
J Biol Chem ; 282(14): 10341-51, 2007 Apr 06.
Article in English | MEDLINE | ID: mdl-17287212

ABSTRACT

The AMP-activated protein kinase (AMPK) is a central regulator of the energy status of the cell, based on its unique ability to respond directly to fluctuations in the ratio of AMP:ATP. Because glucose and amino acids stimulate insulin release from pancreatic beta-cells by the regulation of metabolic intermediates, AMPK represents an attractive candidate for control of beta-cell function. Here, we show that inhibition of AMPK in beta-cells by high glucose inversely correlates with activation of the mammalian Target of Rapamycin (mTOR) pathway, another cellular sensor for nutritional conditions. Forced activation of AMPK by AICAR, phenformin, or oligomycin significantly blocked phosphorylation of p70S6K, a downstream target of mTOR, in response to the combination of glucose and amino acids. Amino acids also suppressed the activity of AMPK, and this at a minimum required the presence of leucine and glutamine. It is unlikely that the ability of AMPK to sense both glucose and amino acids plays a role in regulation of insulin secretion, as inhibition of AMPK by amino acids did not influence insulin secretion. Moreover, activation of AMPK by AICAR or phenformin did not antagonize glucose-stimulated insulin secretion, and insulin secretion was also unaffected in response to suppression of AMPK activity by expression of a dominant negative AMPK construct (K45R). Taken together, these results suggest that the inhibition of AMPK activity by glucose and amino acids might be an important component of the mechanism for nutrient-stimulated mTOR activity but not insulin secretion in the beta-cell.


Subject(s)
Glucose/metabolism , Insulin-Secreting Cells/enzymology , Protein Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Amino Acids/metabolism , Animals , Cell Line , Enzyme Activation/drug effects , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Male , Rats , Rats, Wistar , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases
10.
Am J Physiol Endocrinol Metab ; 292(3): E802-11, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17106064

ABSTRACT

The AMP-activated protein kinase (AMPK) is an important metabolic sensor/effector that coordinates many of the changes in mammalian tissues during variations in energy availability. We have sought to create an in vivo genetic model of chronic AMPK activation, selecting murine skeletal muscle as a representative tissue where AMPK plays important roles. Muscle-selective expression of a mutant noncatalytic gamma1 subunit (R70Qgamma) of AMPK activates AMPK and increases muscle glycogen content. The increase in glycogen content requires the presence of the endogenous AMPK catalytic alpha-subunit, since the offspring of cross-breeding of these mice with mice expressing a dominant negative AMPKalpha subunit have normal glycogen content. In R70Qgamma1-expressing mice, there is a small, but significant, increase in muscle glycogen synthase (GSY) activity associated with an increase in the muscle expression of the liver isoform GSY2. The increase in glycogen content is accompanied, as might be expected, by an increase in exercise capacity. Transgene expression of this mutant AMPKgamma1 subunit may provide a useful model for the chronic activation of AMPK in other tissues to clarify its multiple roles in the regulation of metabolism and other physiological processes.


Subject(s)
Glycogen/metabolism , Multienzyme Complexes/metabolism , Muscle, Skeletal/metabolism , Protein Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Animals , Glycogen Synthase/metabolism , Mice , Mice, Inbred Strains , Mice, Transgenic , Models, Genetic , Multienzyme Complexes/genetics , Physical Conditioning, Animal , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Transgenes
11.
J Biol Chem ; 281(48): 36662-72, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17023420

ABSTRACT

The AMP-activated protein kinase (AMPK) and cAMP signaling systems are both key regulators of cellular metabolism. In this study, we show that AMPK activity is attenuated in response to cAMP-elevating agents through modulation of at least two of its alpha subunit phosphorylation sites, viz. alpha-Thr(172) and alpha1-Ser(485)/alpha2-Ser(491), in the clonal beta-cell line INS-1 as well as in mouse embryonic fibroblasts and COS cells. Forskolin, isobutylmethylxanthine, and the glucose-dependent insulinotropic peptide inhibited AMPK activity and reduced phosphorylation of the activation loop alpha-Thr(172) via inhibition of calcium/calmodulin-dependent protein kinase kinase-alpha and -beta, but not LKB1. These agents also enhanced phosphorylation of alpha-Ser(485/491) by the cAMP-dependent protein kinase. AMPK alpha-Ser(485/491) phosphorylation was necessary but not sufficient for inhibition of AMPK activity in response to forskolin/isobutylmethylxanthine. We show that AMPK alpha-Ser(485/491) can be a site for autophosphorylation, which may play a role in limiting AMPK activation in response to energy depletion or other regulators. Thus, our findings not only demonstrate cross-talk between the cAMP/cAMP-dependent protein kinase and AMPK signaling modules, but also describe a novel mechanism by which multisite phosphorylation of AMPK contributes to regulation of its enzyme activity.


Subject(s)
Gene Expression Regulation, Enzymologic , Multienzyme Complexes/physiology , Protein Serine-Threonine Kinases/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , AMP-Activated Protein Kinases , Animals , COS Cells , Calcium-Calmodulin-Dependent Protein Kinase Kinase , Chlorocebus aethiops , Colforsin/pharmacology , Cyclic AMP/metabolism , Glucose/metabolism , Mice , Multienzyme Complexes/metabolism , Peptides/chemistry , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Rats
12.
FEBS J ; 273(12): 2730-48, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16817901

ABSTRACT

Cyclic AMP responsive element (CRE)-binding protein (CREB) is known to activate transcription when its Ser133 is phosphorylated. Two independent investigations have suggested the presence of Ser133-independent activation. One study identified a kinase, salt-inducible kinase (SIK), which repressed CREB; the other isolated a novel CREB-specific coactivator, transducer of regulated CREB activity (TORC), which upregulated CREB activity. These two opposing signals are connected by the fact that SIK phosphorylates TORC and induces its nuclear export. Because LKB1 has been reported to be an upstream kinase of SIK, we used LKB1-defective HeLa cells to further elucidate TORC-dependent CREB activation. In the absence of LKB1, SIK was unable to phosphorylate TORC, which led to constitutive activation of CRE activity. Overexpression of LKB1 in HeLa cells improved the CRE-dependent transcription in a regulated manner. The inactivation of kinase cascades by 10 nm staurosporine in LKB1-positive HEK293 cells also induced unregulated, constitutively activated, CRE activity. Treatment with staurosporine completely inhibited SIK kinase activity without any significant effect on the phosphorylation level at the LKB1-phosphorylatable site in SIK or the activity of AMPK, another target of LKB1. Constitutive activation of CREB in LKB1-defective cells or in staurosporine-treated cells was not accompanied by CREB phosphorylation at Ser133. The results suggest that LKB1 and its downstream SIK play an important role in silencing CREB activity via the phosphorylation of TORC, and such silencing may be indispensable for the regulated activation of CREB.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , AMP-Activated Protein Kinases , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , COS Cells , Cell Nucleus/metabolism , Chlorocebus aethiops , Cyclic AMP Response Element-Binding Protein/genetics , Cytoplasm/metabolism , Gene Silencing , HeLa Cells , Humans , Mice , Molecular Sequence Data , Multienzyme Complexes/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Rats , Serine/metabolism , Signal Transduction/genetics , Staurosporine/pharmacology , Threonine/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors , Transcription, Genetic , Tumor Cells, Cultured
13.
Trends Biochem Sci ; 31(1): 13-6, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16356723

ABSTRACT

AMP-activated protein kinase (AMPK), a key regulator of energy homeostasis in mammalian cells, is, in turn, regulated by long-sought upstream protein kinases (AMPKKs). Following the recent identification of the tumor-suppressor kinase LKB1 as an AMPKK, a broader role for AMPK in metabolic economy has been unveiled by a new body of work from three groups that implicates the Ca(2+)/calmodulin-dependent protein kinase kinases as AMPKKs. We suggest that PKE (protein kinase "energy" or "economy") is now an apt name for this kinase, which regulates both cellular and whole-organism energy homeostasis.


Subject(s)
Multienzyme Complexes/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases , Calcium-Calmodulin-Dependent Protein Kinase Kinase , Energy Metabolism , Homeostasis , Models, Biological , Models, Molecular , Multienzyme Complexes/chemistry , Protein Conformation , Protein Serine-Threonine Kinases/chemistry
14.
J Biol Chem ; 281(7): 4231-41, 2006 Feb 17.
Article in English | MEDLINE | ID: mdl-16361706

ABSTRACT

AMP-activated kinase (AMPK) is a ubiquitous metabolic sensor that inhibits the cystic fibrosis (CF) transmembrane conductance regulator (CFTR). To determine whether CFTR reciprocally regulates AMPK function in airway epithelia and whether such regulation is involved in lung inflammation, AMPK localization, expression, and activity and cellular metabolic profiles were compared as a function of CFTR status in CF and non-CF primary human bronchial epithelial (HBE) cells. As compared with non-CF HBE cells, CF cells had greater and more diffuse AMPK staining and had greater AMPK activity than their morphologically matched non-CF counterparts. The cellular [AMP]/[ATP] ratio was higher in undifferentiated than in differentiated non-CF cells, which correlated with AMPK activity under these conditions. However, this nucleotide ratio did not predict AMPK activity in differentiating CF cells. Inhibiting channel activity in non-CF cells did not affect AMPK activity or metabolic status, but expressing functional CFTR in CF cells reduced AMPK activity without affecting cellular [AMP]/[ATP]. Therefore, lack of functional CFTR expression and not loss of channel activity in CF cells appears to up-regulate AMPK activity in CF HBE cells, presumably through non-metabolic effects on upstream regulatory pathways. Compared with wild-type CFTR-expressing immortalized CF bronchial epithelial (CFBE) cells, DeltaF508-CFTR-expressing CFBE cells had greater AMPK activity and greater secretion of tumor necrosis factor-alpha and the interleukins IL-6 and IL-8. Further pharmacologic AMPK activation inhibited inflammatory mediator secretion in both wild type- and DeltaF508-expressing cells, suggesting that AMPK activation in CF airway cells is an adaptive response that reduces inflammation. We propose that therapies to activate AMPK in the CF airway may be beneficial in reducing excessive airway inflammation, a major cause of CF morbidity.


Subject(s)
Bronchi/enzymology , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Cystic Fibrosis/enzymology , Gene Expression Regulation, Enzymologic , Inflammation/etiology , Multienzyme Complexes/genetics , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinases , Amino Acid Sequence , Cell Differentiation , Cell Polarity , Epithelial Cells/enzymology , Humans , Inflammation Mediators/physiology , Molecular Sequence Data , Multienzyme Complexes/physiology , Protein Serine-Threonine Kinases/physiology , Up-Regulation
15.
J Biol Chem ; 280(32): 29060-6, 2005 Aug 12.
Article in English | MEDLINE | ID: mdl-15980064

ABSTRACT

The AMP-activated protein kinase (AMPK) is an important regulator of cellular metabolism in response to metabolic stress and to other regulatory signals. AMPK activity is absolutely dependent upon phosphorylation of AMPKalphaThr-172 in its activation loop by one or more AMPK kinases (AMPKKs). The tumor suppressor kinase, LKB1, is a major AMPKK present in a variety of tissues and cells, but several lines of evidence point to the existence of other AMPKKs. We have employed three cell lines deficient in LKB1 to study AMPK regulation and phosphorylation, HeLa, A549, and murine embryo fibroblasts derived from LKB(-/-) mice. In HeLa and A549 cells, mannitol, 2-deoxyglucose, and ionomycin, but not 5-aminoimidazole-4-carboxamide-1-beta-d-ribofuranoside (AICAR), treatment activates AMPK by alphaThr-172 phosphorylation. These responses, as well as the downstream effects of AMPK on the phosphorylation of acetyl-CoA carboxylase, are largely inhibited by the Ca(2+)/ calmodulin-dependent protein kinase kinase (CaMKK) inhibitor, STO-609. AMPKK activity in HeLa cell lysates measured in vitro is totally inhibited by STO-609 with an IC50 comparable with that of the known CaMKK isoforms, CaMKKalpha and CaMKKbeta. Furthermore, 2-deoxyglucose- and ionomycin-stimulated AMPK activity, alphaThr-172 phosphorylation, and acetyl-CoA carboxylase phosphorylation are substantially reduced in HeLa cells transfected with small interfering RNAs specific for CaMKKalpha and CaMKKbeta. Lastly, the activation of AMPK in response to ionomycin and 2-deoxyglucose is not impaired in LKB1(-/-) murine embryo fibroblasts. These data indicate that the CaMKKs function in intact cells as AMPKKs, predicting wider roles for these kinases in regulating AMPK activity in vivo.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Protein Kinases/physiology , AMP-Activated Protein Kinase Kinases , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Benzimidazoles/pharmacology , COS Cells , Cell Line, Tumor , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Deoxyglucose/chemistry , Deoxyglucose/metabolism , Enzyme Activation , Fibroblasts/metabolism , HeLa Cells , Humans , Immunoblotting , Ionomycin/pharmacology , Isoquinolines/pharmacology , Mannitol/chemistry , Mice , Naphthalimides , Phosphorylation , Protein Isoforms , Protein Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Ribonucleotides/pharmacology , Threonine/chemistry
16.
J Biol Chem ; 280(14): 13395-400, 2005 Apr 08.
Article in English | MEDLINE | ID: mdl-15695819

ABSTRACT

AMP-activated protein kinase (AMPK) is an important metabolic stress-sensing protein kinase responsible for regulating metabolism in response to changing energy demand and nutrient supply. Mammalian AMPK is a stable alphabetagamma heterotrimer comprising a catalytic alpha and two non-catalytic subunits, beta and gamma. The beta subunit targets AMPK to membranes via an N-terminal myristoyl group and to glycogen via a mid-molecule glycogen-binding domain. Here we find that the conserved C-terminal 85-residue sequence of the beta subunit, beta1-(186-270), is sufficient to form an active AMP-dependent heterotrimer alpha1beta1-(186-270)-gamma1, whereas the 25-residue beta1 C-terminal (246-270) sequence is sufficient to bind gamma1, gamma2, or gamma3 but not the alpha subunit. Deletion of the beta C-terminal Ile-270 precludes betagamma association in the absence of the alpha subunit, but the presence of the alpha subunit or substitution of Ile-270 with Ala or Glu restores betagamma binding. Truncation of the alpha subunit reveals that beta1 binding requires the alpha1-(313-473) sequence. The conserved C-terminal 85-residue sequence of the beta subunit (90% between beta1 and beta2) is the primary alphagamma binding sequence responsible for the formation of the AMPK alphabetagamma heterotrimer.


Subject(s)
Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Quaternary , Protein Subunits/metabolism , AMP-Activated Protein Kinases , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Fungal Proteins/genetics , Fungal Proteins/metabolism , Humans , Molecular Sequence Data , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Protein Subunits/genetics , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment
17.
Circ Res ; 96(3): 337-45, 2005 Feb 18.
Article in English | MEDLINE | ID: mdl-15653571

ABSTRACT

AMP-activated protein kinase (AMPK) is emerging as an important signaling protein during myocardial ischemia. AMPK is a heterotrimeric complex containing an alpha catalytic subunit and beta and gamma regulatory subunits. Phosphorylation of Thr172 in the activation loop of the alpha subunit by upstream AMPK kinase(s) (AMPKK) is a critical determinant of AMPK activity. However, the mechanisms regulating AMPK phosphorylation in the ischemic heart remain uncertain and were therefore investigated. In the isolated working rat heart, low-flow ischemia rapidly activated AMPKK activity when measured using recombinant AMPK (rAMPK) as substrate. The addition of AMP (10 to 200 micromol/L) augmented the ability of heterotrimeric alpha1beta1gamma1 or alpha2beta1gamma1 rAMPK to be phosphorylated by heart AMPKK in vitro, whereas physiologic concentrations of ATP inhibited rAMPK phosphorylation. However, neither AMP nor ATP directly influenced AMPKK activity: they had no effect on AMPKK-mediated phosphorylation of rAMPK substrates lacking normal AMP-binding gamma subunits (isolated truncated alpha1(1-312) or alpha1beta1gamma1 rAMPK containing an R70Q mutation in the gamma1 AMP-binding site). Regional ischemia in vivo also increased AMPKK activity and AMPK phosphorylation in the rat heart. AMPK phosphorylation could also be induced in vivo without activating AMPKK: AICAR infusion increased AMPK phosphorylation without activating AMPKK; however, the AMP-mimetic AICAR metabolite ZMP enhanced the ability of heterotrimeric rAMPK to be phosphorylated by AMPKK. Thus, heart AMPKK activity is increased by ischemia and its ability to phosphorylate AMPK is highly modulated by the interaction of AMP and ATP with the heterotrimeric AMPK complex, indicating that dual mechanisms regulate AMPKK action in the ischemic heart.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Myocardial Ischemia/enzymology , Protein Kinases/metabolism , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases , Adenosine Monophosphate/pharmacology , Adenosine Triphosphate/pharmacology , Aminoimidazole Carboxamide/metabolism , Aminoimidazole Carboxamide/pharmacology , Animals , Infusions, Intravenous , Male , Multienzyme Complexes/metabolism , Myocardium/enzymology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Ribonucleotides/metabolism , Ribonucleotides/pharmacology
18.
Genes Dev ; 18(23): 2893-904, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15545625

ABSTRACT

Mammalian target of rapamycin (mTOR) is a central regulator of protein synthesis whose activity is modulated by a variety of signals. Energy depletion and hypoxia result in mTOR inhibition. While energy depletion inhibits mTOR through a process involving the activation of AMP-activated protein kinase (AMPK) by LKB1 and subsequent phosphorylation of TSC2, the mechanism of mTOR inhibition by hypoxia is not known. Here we show that mTOR inhibition by hypoxia requires the TSC1/TSC2 tumor suppressor complex and the hypoxia-inducible gene REDD1/RTP801. Disruption of the TSC1/TSC2 complex through loss of TSC1 or TSC2 blocks the effects of hypoxia on mTOR, as measured by changes in the mTOR targets S6K and 4E-BP1, and results in abnormal accumulation of Hypoxia-inducible factor (HIF). In contrast to energy depletion, mTOR inhibition by hypoxia does not require AMPK or LKB1. Down-regulation of mTOR activity by hypoxia requires de novo mRNA synthesis and correlates with increased expression of the hypoxia-inducible REDD1 gene. Disruption of REDD1 abrogates the hypoxia-induced inhibition of mTOR, and REDD1 overexpression is sufficient to down-regulate S6K phosphorylation in a TSC1/TSC2-dependent manner. Inhibition of mTOR function by hypoxia is likely to be important for tumor suppression as TSC2-deficient cells maintain abnormally high levels of cell proliferation under hypoxia.


Subject(s)
Hypoxia/physiopathology , Protein Kinases/physiology , Repressor Proteins/physiology , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , 3T3 Cells , Animals , Cell Division/physiology , Down-Regulation , Mice , Mice, Inbred C57BL , Phosphorylation , RNA, Small Interfering , Ribosomal Protein S6 Kinases/metabolism , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein
19.
Proc Natl Acad Sci U S A ; 101(10): 3329-35, 2004 Mar 09.
Article in English | MEDLINE | ID: mdl-14985505

ABSTRACT

AMP-activated protein kinase (AMPK) is a highly conserved sensor of cellular energy status found in all eukaryotic cells. AMPK is activated by stimuli that increase the cellular AMP/ATP ratio. Essential to activation of AMPK is its phosphorylation at Thr-172 by an upstream kinase, AMPKK, whose identity in mammalian cells has remained elusive. Here we present biochemical and genetic evidence indicating that the LKB1 serine/threonine kinase, the gene inactivated in the Peutz-Jeghers familial cancer syndrome, is the dominant regulator of AMPK activation in several mammalian cell types. We show that LKB1 directly phosphorylates Thr-172 of AMPKalpha in vitro and activates its kinase activity. LKB1-deficient murine embryonic fibroblasts show nearly complete loss of Thr-172 phosphorylation and downstream AMPK signaling in response to a variety of stimuli that activate AMPK. Reintroduction of WT, but not kinase-dead, LKB1 into these cells restores AMPK activity. Furthermore, we show that LKB1 plays a biologically significant role in this pathway, because LKB1-deficient cells are hypersensitive to apoptosis induced by energy stress. On the basis of these results, we propose a model to explain the apparent paradox that LKB1 is a tumor suppressor, yet cells lacking LKB1 are resistant to cell transformation by conventional oncogenes and are sensitive to killing in response to agents that elevate AMP. The role of LKB1/AMPK in the survival of a subset of genetically defined tumor cells may provide opportunities for cancer therapeutics.


Subject(s)
Apoptosis/physiology , Carrier Proteins , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteins , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Cell Line , Energy Metabolism , Enzyme Activation , HeLa Cells , Humans , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , LLC-PK1 Cells , Mice , Mice, Knockout , Models, Biological , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Phosphorylation , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Swine , Threonine/chemistry , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
20.
Protein Sci ; 13(1): 155-65, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14691231

ABSTRACT

AMP-activated protein kinase (AMPK) is a alphabetagamma heterotrimer that is activated in response to both hormones and intracellular metabolic stress signals. AMPK is regulated by phosphorylation on the alpha subunit and by AMP allosteric control previously thought to be mediated by both alpha and gamma subunits. Here we present evidence that adjacent gamma subunit pairs of CBS repeat sequences (after Cystathionine Beta Synthase) form an AMP binding site related to, but distinct from the classical AMP binding site in phosphorylase, that can also bind ATP. The AMP binding site of the gamma(1) CBS1/CBS2 pair, modeled on the structures of the CBS sequences present in the inosine monophosphate dehydrogenase crystal structure, contains three arginine residues 70, 152, and 171 and His151. The yeast gamma homolog, snf4 contains a His151Gly substitution, and when this is introduced into gamma(1), AMP allosteric control is substantially lost and explains why the yeast snf1p/snf4p complex is insensitive to AMP. Arg70 in gamma(1) corresponds to the site of mutation in human gamma(2) and pig gamma(3) genes previously identified to cause an unusual cardiac phenotype and glycogen storage disease, respectively. Mutation of any of AMP binding site Arg residues to Gln substantially abolishes AMP allosteric control in expressed AMPK holoenzyme. The Arg/Gln mutations also suppress the previously described inhibitory properties of ATP and render the enzyme constitutively active. We propose that ATP acts as an intrasteric inhibitor by bridging the alpha and gamma subunits and that AMP functions to derepress AMPK activity.


Subject(s)
Adenosine Monophosphate/metabolism , Allosteric Site , Protein Kinases/chemistry , Protein Kinases/metabolism , Protein Subunits/chemistry , Protein Subunits/metabolism , AMP-Activated Protein Kinase Kinases , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Conserved Sequence , Enzyme Activation , Glycine/metabolism , Hydrogen Bonding , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Point Mutation , Protein Binding , Protein Kinases/genetics , Protein Structure, Tertiary , Protein Subunits/genetics , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...