Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
EMBO J ; 38(24): e102578, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31381180

ABSTRACT

Exercise stimulates cellular and physiological adaptations that are associated with widespread health benefits. To uncover conserved protein phosphorylation events underlying this adaptive response, we performed mass spectrometry-based phosphoproteomic analyses of skeletal muscle from two widely used rodent models: treadmill running in mice and in situ muscle contraction in rats. We overlaid these phosphoproteomic signatures with cycling in humans to identify common cross-species phosphosite responses, as well as unique model-specific regulation. We identified > 22,000 phosphosites, revealing orthologous protein phosphorylation and overlapping signaling pathways regulated by exercise. This included two conserved phosphosites on stromal interaction molecule 1 (STIM1), which we validate as AMPK substrates. Furthermore, we demonstrate that AMPK-mediated phosphorylation of STIM1 negatively regulates store-operated calcium entry, and this is beneficial for exercise in Drosophila. This integrated cross-species resource of exercise-regulated signaling in human, mouse, and rat skeletal muscle has uncovered conserved networks and unraveled crosstalk between AMPK and intracellular calcium flux.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Calcium Channels/metabolism , Calcium/metabolism , Proteomics/methods , Stromal Interaction Molecule 1/metabolism , Animals , Calcium Signaling/physiology , Drosophila , Female , Humans , Male , Membrane Proteins , Mice , Muscle, Skeletal/metabolism , Phosphorylation , Protein Conformation , Rats , Rats, Wistar , Signal Transduction , Stromal Interaction Molecule 1/chemistry , Stromal Interaction Molecule 1/genetics
2.
Biochem J ; 479(11): 1181-1204, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35552369

ABSTRACT

The AMP-activated protein kinase (AMPK) αßγ heterotrimer is a primary cellular energy sensor and central regulator of energy homeostasis. Activating skeletal muscle AMPK with small molecule drugs improves glucose uptake and provides an opportunity for new strategies to treat type 2 diabetes and insulin resistance, with recent genetic and pharmacological studies indicating the α2ß2γ1 isoform combination as the heterotrimer complex primarily responsible. With the goal of developing α2ß2-specific activators, here we perform structure/function analysis of the 2-hydroxybiphenyl group of SC4, an activator with tendency for α2-selectivity that is also capable of potently activating ß2 complexes. Substitution of the LHS 2-hydroxyphenyl group with polar-substituted cyclohexene-based probes resulted in two AMPK agonists, MSG010 and MSG011, which did not display α2-selectivity when screened against a panel of AMPK complexes. By radiolabel kinase assay, MSG010 and MSG011 activated α2ß2γ1 AMPK with one order of magnitude greater potency than the pan AMPK activator MK-8722. A crystal structure of MSG011 complexed to AMPK α2ß1γ1 revealed a similar binding mode to SC4 and the potential importance of an interaction between the SC4 2-hydroxyl group and α2-Lys31 for directing α2-selectivity. MSG011 induced robust AMPK signalling in mouse primary hepatocytes and commonly used cell lines, and in most cases this occurred in the absence of changes in phosphorylation of the kinase activation loop residue α-Thr172, a classical marker of AMP-induced AMPK activity. These findings will guide future design of α2ß2-selective AMPK activators, that we hypothesise may avoid off-target complications associated with indiscriminate activation of AMPK throughout the body.


Subject(s)
AMP-Activated Protein Kinases , Diabetes Mellitus, Type 2 , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Diabetes Mellitus, Type 2/metabolism , Mice , Muscle, Skeletal/metabolism , Phosphorylation
3.
J Biol Chem ; 295(48): 16239-16250, 2020 11 27.
Article in English | MEDLINE | ID: mdl-32913128

ABSTRACT

The calcium-calmodulin-dependent protein kinase kinase-2 (CaMKK2) is a key regulator of cellular and whole-body energy metabolism. It is known to be activated by increases in intracellular Ca2+, but the mechanisms by which it is inactivated are less clear. CaMKK2 inhibition protects against prostate cancer, hepatocellular carcinoma, and metabolic derangements induced by a high-fat diet; therefore, elucidating the intracellular mechanisms that inactivate CaMKK2 has important therapeutic implications. Here we show that stimulation of cAMP-dependent protein kinase A (PKA) signaling in cells inactivates CaMKK2 by phosphorylation of three conserved serine residues. PKA-dependent phosphorylation of Ser495 directly impairs calcium-calmodulin activation, whereas phosphorylation of Ser100 and Ser511 mediate recruitment of 14-3-3 adaptor proteins that hold CaMKK2 in the inactivated state by preventing dephosphorylation of phospho-Ser495 We also report the crystal structure of 14-3-3ζ bound to a synthetic diphosphorylated peptide that reveals how the canonical (Ser511) and noncanonical (Ser100) 14-3-3 consensus sites on CaMKK2 cooperate to bind 14-3-3 proteins. Our findings provide detailed molecular insights into how cAMP-PKA signaling inactivates CaMKK2 and reveals a pathway to inhibit CaMKK2 with potential for treating human diseases.


Subject(s)
14-3-3 Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Signal Transduction , 14-3-3 Proteins/genetics , Animals , COS Cells , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Cell Line, Tumor , Chlorocebus aethiops , Cyclic AMP-Dependent Protein Kinases/genetics , Enzyme Activation , Humans
4.
J Cell Sci ; 132(7)2019 04 03.
Article in English | MEDLINE | ID: mdl-30814334

ABSTRACT

AMP-activated kinase (AMPK) and target of rapamycin (TOR) signalling coordinate cell growth, proliferation, metabolism and cell survival with the nutrient environment of cells. The poor vasculature and nutritional stress experienced by cells in solid tumours raises the question: how do they assimilate sufficient nutrients to survive? Here, we show that human and fission yeast cells import ATP and AMP from their external environment to regulate AMPK and TOR signalling. Exposure of fission yeast (Schizosaccharomyces pombe) and human cells to external AMP impeded cell growth; however, in yeast this restraining impact required AMPK. In contrast, external ATP rescued the growth defect of yeast mutants with reduced TORC1 signalling; furthermore, exogenous ATP transiently enhanced TORC1 signalling in both yeast and human cell lines. Addition of the PANX1 channel inhibitor probenecid blocked ATP import into human cell lines suggesting that this channel may be responsible for both ATP release and uptake in mammals. In light of these findings, it is possible that the higher extracellular ATP concentration reported in solid tumours is both scavenged and recognized as an additional energy source beneficial for cell growth.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Signal Transduction , AMP-Activated Protein Kinases/genetics , Cell Proliferation , Connexins/metabolism , Gene Expression Regulation, Fungal , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1/genetics , Nerve Tissue Proteins/metabolism , Phosphorylation , Schizosaccharomyces , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Stress, Physiological
5.
Int J Mol Sci ; 22(3)2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33513781

ABSTRACT

Physical exercise elicits physiological metabolic perturbations such as energetic and oxidative stress; however, a diverse range of cellular processes are stimulated in response to combat these challenges and maintain cellular energy homeostasis. AMP-activated protein kinase (AMPK) is a highly conserved enzyme that acts as a metabolic fuel sensor and is central to this adaptive response to exercise. The complexity of AMPK's role in modulating a range of cellular signalling cascades is well documented, yet aside from its well-characterised regulation by activation loop phosphorylation, AMPK is further subject to a multitude of additional regulatory stimuli. Therefore, in this review we comprehensively outline current knowledge around the post-translational modifications of AMPK, including novel phosphorylation sites, as well as underappreciated roles for ubiquitination, sumoylation, acetylation, methylation and oxidation. We provide insight into the physiological ramifications of these AMPK modifications, which not only affect its activity, but also subcellular localisation, nutrient interactions and protein stability. Lastly, we highlight the current knowledge gaps in this area of AMPK research and provide perspectives on how the field can apply greater rigour to the characterisation of novel AMPK regulatory modifications.


Subject(s)
AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/metabolism , Energy Metabolism , Protein Processing, Post-Translational , AMP-Activated Protein Kinases/genetics , Acetylation , Animals , Homeostasis , Humans , Methylation , Oxidation-Reduction , Oxidative Stress , Phosphorylation , Protein Domains , Signal Transduction/genetics , Ubiquitination
6.
Int J Mol Sci ; 22(8)2021 Apr 14.
Article in English | MEDLINE | ID: mdl-33919972

ABSTRACT

As life expectancy has increased, particularly in developed countries, due to medical advances and increased prosperity, age-related neurological diseases and mental health disorders have become more prevalent health issues, reducing the well-being and quality of life of sufferers and their families. In recent decades, due to reduced work-related levels of physical activity, and key research insights, prescribing adequate exercise has become an innovative strategy to prevent or delay the onset of these pathologies and has been demonstrated to have therapeutic benefits when used as a sole or combination treatment. Recent evidence suggests that the beneficial effects of exercise on the brain are related to several underlying mechanisms related to muscle-brain, liver-brain and gut-brain crosstalk. Therefore, this review aims to summarize the most relevant current knowledge of the impact of exercise on mood disorders and neurodegenerative diseases, and to highlight the established and potential underlying mechanisms involved in exercise-brain communication and their benefits for physiology and brain function.


Subject(s)
Brain/physiology , Exercise/physiology , Gastrointestinal Microbiome/physiology , Nervous System Diseases/therapy , Humans , Nervous System Diseases/microbiology , Nervous System Diseases/physiopathology , Quality of Life
7.
FASEB J ; 33(12): 14825-14840, 2019 12.
Article in English | MEDLINE | ID: mdl-31670977

ABSTRACT

ATPase inhibitory factor 1 (IF1) is an ATP synthase-interacting protein that suppresses the hydrolysis activity of ATP synthase. In this study, we observed that the expression of IF1 was up-regulated in response to electrical pulse stimulation of skeletal muscle cells and in exercized mice and healthy men. IF1 stimulates glucose uptake via AMPK in skeletal muscle cells and primary cultured myoblasts. Reactive oxygen species and Rac family small GTPase 1 (Rac1) function in the upstream and downstream of AMPK, respectively, in IF1-mediated glucose uptake. In diabetic animal models, the administration of recombinant IF1 improved glucose tolerance and down-regulated blood glucose level. In addition, IF1 inhibits ATP hydrolysis by ß-F1-ATPase in plasma membrane, thereby increasing extracellular ATP and activating the protein kinase B (Akt) pathway, ultimately leading to glucose uptake. Thus, we suggest that IF1 is a novel myokine and propose a mechanism by which AMPK and Akt contribute independently to IF1-mediated improvement of glucose tolerance impairment. These results demonstrate the importance of IF1 as a potential antidiabetic agent.-Lee, H. J., Moon, J., Chung, I., Chung, J. H., Park, C., Lee, J. O., Han, J. A., Kang, M. J., Yoo, E. H., Kwak, S.-Y., Jo, G., Park, W., Park, J., Kim, K. M., Lim, S., Ngoei, K. R. W., Ling, N. X. Y., Oakhill, J. S., Galic, S., Murray-Segal, L., Kemp, B. E., Mantzoros, C. S., Krauss, R. M., Shin, M.-J., Kim, H. S. ATP synthase inhibitory factor 1 (IF1), a novel myokine, regulates glucose metabolism by AMPK and Akt dual pathways.


Subject(s)
Glucose/metabolism , Myoblasts/metabolism , Proteins/metabolism , AMP-Activated Protein Kinase Kinases , Adenosine Triphosphate/metabolism , Adult , Animals , Cell Line , Cells, Cultured , Diabetes Mellitus, Type 2/drug therapy , Humans , Hypoglycemic Agents/therapeutic use , Male , Mice , Mice, Inbred C57BL , Protein Kinases/metabolism , Proteins/genetics , Proteins/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/therapeutic use , ATPase Inhibitory Protein
8.
Bipolar Disord ; 22(8): 841-848, 2020 12.
Article in English | MEDLINE | ID: mdl-32216002

ABSTRACT

OBJECTIVES: Loss-of-function mutations in the gene encoding the calcium-calmodulin (Ca2+ -CaM)-dependent protein kinase kinase-2 (CaMKK2) enzyme are linked to bipolar disorder. Recently, a de novo arginine to cysteine (R311C) mutation in CaMKK2 was identified from a whole exome sequencing study of bipolar patients and their unaffected parents. The aim of the present study was to determine the functional consequences of the R311C mutation on CaMKK2 activity and regulation by Ca2+ -CaM. METHODS: The effects of the R311C mutation on CaMKK2 activity and Ca2+ -CaM activation were examined using a radiolabeled adenosine triphosphate (ATP) kinase assay. We performed immunoblot analysis to determine whether the R311C mutation impacts threonine-85 (T85) autophosphorylation, an activating phosphorylation site on CaMKK2 that has also been implicated in bipolar disorder. We also expressed the R311C mutant in CaMKK2 knockout HAP1 cells and used immunoblot analysis and an MTS reduction assay to study its effects on Ca2+ -dependent downstream signaling and cell viability, respectively. RESULTS: The R311C mutation maps to the conserved HRD motif within the catalytic loop of CaMKK2 and caused a marked reduction in kinase activity and Ca2+ -CaM activation. The R311C mutation virtually abolished T85 autophosphorylation in response to Ca2+ -CaM and exerted a dominant-negative effect in cells as it impaired the ability of wild-type CaMKK2 to initiate downstream signaling and maintain cell viability. CONCLUSIONS: The highly disruptive, loss-of-function impact of the de novo R311C mutation in human CaMKK2 provides a compelling functional rationale for being considered a potential rare monogenic cause of bipolar disorder.


Subject(s)
Bipolar Disorder/genetics , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Calcium/metabolism , Calmodulin/metabolism , Bipolar Disorder/diagnosis , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Calmodulin/genetics , Genetic Variation , Humans , Mutation , Phosphorylation , Signal Transduction/physiology
9.
Biochem J ; 476(12): 1725-1740, 2019 06 26.
Article in English | MEDLINE | ID: mdl-31189568

ABSTRACT

AMP-activated protein kinase (AMPK) is a heterotrimer of α-catalytic and ß- and γ-regulatory subunits that acts to regulate cellular and whole-body nutrient metabolism. The key role of AMPK in sensing energy status has led to significant interest in AMPK as a therapeutic target for dysfunctional metabolism in type 2 diabetes, insulin resistance and obesity. Despite the actions of AMPK in the liver and skeletal muscle being extensively studied, the role of AMPK in adipose tissue and adipocytes remains less well characterised. Small molecules that selectively influence AMPK heterotrimers containing specific AMPKß subunit isoforms have been developed, including MT47-100, which selectively inhibits complexes containing AMPKß2. AMPKß1 and AMPKß2 are the principal AMPKß subunit isoforms in rodent liver and skeletal muscle, respectively, yet the contribution of specific AMPKß isoforms to adipose tissue function, however, remains largely unknown. This study therefore sought to determine the contribution of AMPKß subunit isoforms to adipocyte biology, focussing on adipogenesis. AMPKß2 was the principal AMPKß isoform in 3T3-L1 adipocytes, isolated rodent adipocytes and human subcutaneous adipose tissue, as assessed by the contribution to total cellular AMPK activity. Down-regulation of AMPKß2 with siRNA inhibited lipid accumulation, cellular adiponectin levels and adiponectin secretion during 3T3-L1 adipogenesis, whereas down-regulation of AMPKß1 had no effect. Incubation of 3T3-L1 cells with MT47-100 selectively inhibited AMPK complexes containing AMPKß2 whilst simultaneously inhibiting cellular lipid accumulation as well as cellular levels and secretion of adiponectin. Taken together, these data indicate that increased expression of AMPKß2 is an important feature of efficient adipogenesis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipocytes/enzymology , Adipogenesis , Gene Expression Regulation, Enzymologic , Lipid Metabolism , Up-Regulation , 3T3-L1 Cells , Animals , Humans , Isoenzymes/metabolism , Male , Mice , Rats, Sprague-Dawley
10.
Molecules ; 25(2)2020 Jan 13.
Article in English | MEDLINE | ID: mdl-31941153

ABSTRACT

The calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) activates CAMK1, CAMK4, AMPK, and AKT, leading to numerous physiological responses. The deregulation of CAMKK2 is linked to several diseases, suggesting the utility of CAMKK2 inhibitors for oncological, metabolic and inflammatory indications. In this work, we demonstrate that STO-609, frequently described as a selective inhibitor for CAMKK2, potently inhibits a significant number of other kinases. Through an analysis of literature and public databases, we have identified other potent CAMKK2 inhibitors and verified their activities in differential scanning fluorimetry and enzyme inhibition assays. These inhibitors are potential starting points for the development of selective CAMKK2 inhibitors and will lead to tools that delineate the roles of this kinase in disease biology.


Subject(s)
Benzimidazoles/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Kinase , Naphthalimides/chemistry , Protein Kinase Inhibitors/chemistry , Animals , Calcium-Calmodulin-Dependent Protein Kinase Kinase/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Kinase/chemistry , Humans
11.
J Biol Chem ; 293(23): 8874-8885, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29695504

ABSTRACT

Inhibition of the metabolic regulator AMP-activated protein kinase (AMPK) is increasingly being investigated for its therapeutic potential in diseases where AMPK hyperactivity results in poor prognoses, as in established cancers and neurodegeneration. However, AMPK-inhibitory tool compounds are largely limited to compound C, which has a poor selectivity profile. Here we identify the pyrimidine derivative SBI-0206965 as a direct AMPK inhibitor. SBI-0206965 inhibits AMPK with 40-fold greater potency and markedly lower kinase promiscuity than compound C and inhibits cellular AMPK signaling. Biochemical characterization reveals that SBI-0206965 is a mixed-type inhibitor. A co-crystal structure of the AMPK kinase domain/SBI-0206965 complex shows that the drug occupies a pocket that partially overlaps the ATP active site in a type IIb inhibitor manner. SBI-0206965 has utility as a tool compound for investigating physiological roles for AMPK and provides fresh impetus to small-molecule AMPK inhibitor therapeutic development.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , Benzamides/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/metabolism , Animals , Benzamides/chemistry , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , Molecular Docking Simulation , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemistry
13.
Biochem Soc Trans ; 47(2): 733-741, 2019 04 30.
Article in English | MEDLINE | ID: mdl-31000529

ABSTRACT

The AMP (adenosine 5'-monophosphate)-activated protein kinase (AMPK) is a key regulator of cellular and whole-body energy homeostasis that co-ordinates metabolic processes to ensure energy supply meets demand. At the cellular level, AMPK is activated by metabolic stresses that increase AMP or adenosine 5'-diphosphate (ADP) coupled with falling adenosine 5'-triphosphate (ATP) and acts to restore energy balance by choreographing a shift in metabolism in favour of energy-producing catabolic pathways while inhibiting non-essential anabolic processes. AMPK also regulates systemic energy balance and is activated by hormones and nutritional signals in the hypothalamus to control appetite and body weight. Failure to maintain energy balance plays an important role in chronic diseases such as obesity, type 2 diabetes and inflammatory disorders, which has prompted a major drive to develop pharmacological activators of AMPK. An array of small-molecule allosteric activators has now been developed, several of which can activate AMPK by direct allosteric activation, independently of Thr172 phosphorylation, which was previously regarded as indispensable for AMPK activity. In this review, we summarise the state-of-the-art regarding our understanding of the molecular mechanisms that govern direct allosteric activation of AMPK by adenylate nucleotides and small-molecule drugs.


Subject(s)
AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Adenosine Diphosphate/metabolism , Adenosine Monophosphate/genetics , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Allosteric Regulation/genetics , Allosteric Regulation/physiology , Animals , Energy Metabolism/genetics , Energy Metabolism/physiology , Humans , Phosphorylation
15.
Biochem J ; 474(7): 1289-1292, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28336748

ABSTRACT

Maintaining a steady balance between nutrient supply and energy demand is essential for all living organisms and is achieved through the dynamic control of metabolic processes that produce and consume adenosine-5'-triphosphate (ATP), the universal currency of energy in all cells. A key sensor of cellular energy is the adenosine-5'-monophosphate (AMP)-activated protein kinase (AMPK), which is the core component of a signaling network that regulates energy and nutrient metabolism. AMPK is activated by metabolic stresses that decrease cellular ATP, and functions to restore energy balance by orchestrating a switch in metabolism away from anabolic pathways toward energy-generating catabolic processes. A new study published in a recent issue of Biochemical Journal by Zibrova et al. shows that glutamine:fructose-6-phosphate amidotransferase-1 (GFAT1), the rate-limiting enzyme of the hexosamine biosynthesis pathway (HBP), is a physiological substrate of AMPK. The HBP is an offshoot of the glycolytic pathway that drives the synthesis of uridine-5'-diphospho-N-acetylglucosamine, the requisite donor metabolite needed for dynamic ß-N-acetylglucosamine (O-GlcNAc) modification (O-GlcNAcylation) of cellular proteins. O-GlcNAcylation is a nutrient-sensitive post-translational modification that, like phosphorylation, regulates numerous intracellular processes. Zibrova et al. show that inhibitory phosphorylation of the GFAT1 residue Ser243 by AMPK in response to physiological or small-molecule activators leads to a reduction in cellular protein O-GlcNAcylation. Further work revealed that AMPK-dependent phosphorylation of GFAT1 promotes angiogenesis in endothelial cells. This elegant study demonstrates that the AMPK-GFAT1 signaling axis serves as an important communication point between two nutrient-sensitive signaling pathways and is likely to play a significant role in controlling physiological processes in many other tissues.


Subject(s)
Acetylglucosamine/metabolism , Endothelial Cells/metabolism , Energy Metabolism/genetics , Protein Processing, Post-Translational , Acylation , Cell Line , Endothelial Cells/cytology , Hexosamines/biosynthesis , Humans , Neovascularization, Physiologic/genetics , Phosphorylation , Signal Transduction
16.
Molecules ; 23(5)2018 May 19.
Article in English | MEDLINE | ID: mdl-29783765

ABSTRACT

We demonstrate for the first time that 4H-1,2,6-thiadiazin-4-one (TDZ) can function as a chemotype for the design of ATP-competitive kinase inhibitors. Using insights from a co-crystal structure of a 3,5-bis(arylamino)-4H-1,2,6-thiadiazin-4-one bound to calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2), several analogues were identified with micromolar activity through targeted displacement of bound water molecules in the active site. Since the TDZ analogues showed reduced promiscuity compared to their 2,4-dianilinopyrimidine counter parts, they represent starting points for development of highly selective kinase inhibitors.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Kinase/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Thiadiazoles/chemical synthesis , Thiadiazoles/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Kinase/chemistry , Catalytic Domain , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Protein Kinase Inhibitors/chemistry , Thiadiazoles/chemistry , Water/chemistry
17.
Plant J ; 82(2): 183-92, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25736509

ABSTRACT

SNF1-related protein kinase 1 (SnRK1) is the plant orthologue of the evolutionarily-conserved SNF1/AMPK/SnRK1 protein kinase family that contributes to cellular energy homeostasis. Functional as heterotrimers, family members comprise a catalytic α subunit and non-catalytic ß and γ subunits; multiple isoforms of each subunit type exist, giving rise to various isoenzymes. The Arabidopsis thaliana genome contains homologues of each subunit type, and, in addition, two atypical subunits, ß(3) and ßγ, with unique domain architecture, that are found only amongst plants, suggesting atypical heterotrimers. The AtSnRK1 subunit structure was determined using recombinant protein expression and endogenous co-immunoprecipitation, and six unique isoenzyme combinations were identified. Each heterotrimeric isoenzyme comprises a catalytic α subunit together with the unique ßγ subunit and one of three non-catalytic ß subunits: ß(1), ß(2) or the plant-specific ß(3) isoform. Thus, the AtSnRK1 heterotrimers contain the atypical ßγ subunit rather than a conventional γ subunit. Mammalian AMPK heterotrimers are phosphorylated on the T-loop (pThr175/176) within both catalytic a subunits. However, AtSnRK1 is insensitive to AMP and ADP, and is resistant to T-loop dephosphorylation by protein phosphatases, a process that inactivates other SNF1/AMPK family members. In addition, we show that SnRK1 is inhibited by a heat-labile, >30 kDa, soluble proteinaceous factor that is present in the lysate of young rosette leaves. Finally, none of the three SnRK1 carbohydrate-binding modules, located in the ß(1), ß(2) and ßγ subunits, associate with various carbohydrates, including starch, the plant analogue of glycogen to which AMPK binds in vitro. These data clearly demonstrate that AtSnRK1 is an atypical member of the SNF1/AMPK/SnRK1 family.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , AMP-Activated Protein Kinases/genetics , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Gene Expression Regulation, Plant/genetics , Gene Expression Regulation, Plant/physiology , Immunoprecipitation , Isoenzymes/genetics , Isoenzymes/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
18.
Biochem J ; 468(1): 125-32, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25742316

ABSTRACT

Metformin is the mainstay therapy for type 2 diabetes (T2D) and many patients also take salicylate-based drugs [i.e., aspirin (ASA)] for cardioprotection. Metformin and salicylate both increase AMP-activated protein kinase (AMPK) activity but by distinct mechanisms, with metformin altering cellular adenylate charge (increasing AMP) and salicylate interacting directly at the AMPK ß1 drug-binding site. AMPK activation by both drugs results in phosphorylation of ACC (acetyl-CoA carboxylase; P-ACC) and inhibition of acetyl-CoA carboxylase (ACC), the rate limiting enzyme controlling fatty acid synthesis (lipogenesis). We find doses of metformin and salicylate used clinically synergistically activate AMPK in vitro and in vivo, resulting in reduced liver lipogenesis, lower liver lipid levels and improved insulin sensitivity in mice. Synergism occurs in cell-free assays and is specific for the AMPK ß1 subunit. These effects are also observed in primary human hepatocytes and patients with dysglycaemia exhibit additional improvements in a marker of insulin resistance (proinsulin) when treated with ASA and metformin compared with either drug alone. These data indicate that metformin-salicylate combination therapy may be efficacious for the treatment of non-alcoholic fatty liver disease (NAFLD) and T2D.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aspirin/administration & dosage , Liver/drug effects , Liver/metabolism , Metformin/administration & dosage , Animals , Cardiotonic Agents/administration & dosage , Cells, Cultured , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Drug Synergism , Enzyme Activation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hypoglycemic Agents/administration & dosage , Insulin Resistance , Lipogenesis/drug effects , Male , Mice , Mice, Inbred C57BL
19.
Biochem Biophys Res Commun ; 443(2): 435-40, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24332941

ABSTRACT

The AMP-activated protein kinase (AMPK) regulates cellular and whole-body energy balance in response to changes in adenylate charge and hormonal signals. Activation of AMPK in tissues such as skeletal muscle and liver reverses many of the metabolic defects associated with obesity and Type 2 diabetes. Here we report a bi-quinoline (JJO-1) that allosterically activates all AMPK αßγ isoforms in vitro except complexes containing the γ3 subunit. JJO-1 does not directly activate the autoinhibited α subunit kinase domain and differs among other known direct activators of AMPK in that allosteric activation occurs only at low ATP concentrations, and is not influenced by either mutation of the γ subunit adenylate-nucleotide binding sites or deletion of the ß subunit carbohydrate-binding module. Our findings indicate that AMPK has multiple modes of allosteric activation that may be exploited to design isoform-specific activators as potential therapeutics for metabolic diseases.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Quinolines/pharmacology , Signal Transduction/physiology , Animals , COS Cells , Chlorocebus aethiops , Enzyme Activation/drug effects , Signal Transduction/drug effects
20.
Essays Biochem ; 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38994736

ABSTRACT

Metabolic homeostasis and the ability to link energy supply to demand are essential requirements for all living cells to grow and proliferate. Key to metabolic homeostasis in all eukaryotes are AMPK and mTORC1, two kinases that sense nutrient levels and function as counteracting regulators of catabolism (AMPK) and anabolism (mTORC1) to control cell survival, growth and proliferation. Discoveries beginning in the early 2000s revealed that AMPK and mTORC1 communicate, or cross-talk, through direct and indirect phosphorylation events to regulate the activities of each other and their shared protein substrate ULK1, the master initiator of autophagy, thereby allowing cellular metabolism to rapidly adapt to energy and nutritional state. More recent reports describe divergent mechanisms of AMPK/mTORC1 cross-talk and the elaborate means by which AMPK and mTORC1 are activated at the lysosome. Here, we provide a comprehensive overview of current understanding in this exciting area and comment on new evidence showing mTORC1 feedback extends to the level of the AMPK isoform, which is particularly pertinent for some cancers where specific AMPK isoforms are implicated in disease pathogenesis.

SELECTION OF CITATIONS
SEARCH DETAIL