Open access

Targeting AMPK for Therapeutic Intervention in Type 2 Diabetes

Written By

Mohamed Kodiha and Ursula Stochaj

Submitted: 10 November 2010 Published: 12 September 2011

DOI: 10.5772/20930

From the Edited Volume

Medical Complications of Type 2 Diabetes

Edited by Colleen Croniger

Chapter metrics overview

4,903 Chapter Downloads

View Full Metrics

1. Introduction

This chapter begins with general information on the role of 5’-AMP activated kinase (AMPK) in human physiology and the molecular mechanisms that control this kinase. We discuss the functions of AMPK in different tissues and their relationship to type 2 diabetes. AMPK substrates in different subcellular organelles and compartments are described, and we speculate how the localized action of AMPK could help to control type 2 diabetes. Our review concludes with future directions that are based on the compartment-specific action of AMPK to develop new therapeutic strategies for patients with type 2 diabetes.

1.1. AMPK activity is critical to cell physiology in different tissues and organs

AMPK functions as a ser/thr kinase which provides an evolutionary conserved cellular energy sensor. This kinase is a focal point for metabolic control in all eukaryotes, where it regulates many aspects of physiology (Hardie, 2008a; Kim et al., 2009a; Li & McCullough, 2010; Lopaschuk, 2008; Ronnett et al., 2009; Steinberg & Kemp, 2009; Zhang, et al., 2009). It is well-established that AMPK and its yeast ortholog Snf1 control a large number of diverse processes; they include the response to nutrient limitation or other environmental changes, transcription, transport across the nuclear envelope, cell growth, cell cycle progression, mitosis, cell polarity, development, auto- and mitophagy (Amato et al., 2011; Bungard et al., 2010; Egan et al., 2010; Lee et al., 2007; Li & McCullough, 2010; Mirouse et al., 2007; Nagata & Hirata, 2009; Narbonne & Roy, 2009; Quan et al., 2007; Steinberg & Kemp, 2009; Viollet et al., 2009a; Wang et al., 2010; Witczak et al., 2008). As a result of these contributions, AMPK is vital to the function of several organs and tissues in metazoans (Fig. 1).

Owing to its pivotal role in the control of glucose homeostasis, carbohydrate, lipid and protein metabolism AMPK is a key player in many human diseases and disorders (Fogarty & Hardie, 2010; Lage et al., 2008; Towler & Hardie, 2007; Viollet et al., 2009b). In particular, the low activation state of AMPK could contribute to the increase in type 2 diabetes and obesity (Hardie et al., 2006). Moreover, as essential regulator of glucose homeostasis and lipid metabolism, AMPK has become an important therapeutic target in type 2 diabetes and obesity. This is exemplified by metformin and thiazolidinedione derivatives (TZDs); these drugs are used for therapeutic intervention in type 2 diabetes and lead to the activation of AMPK.

Figure 1.

The role of AMPK in different organs and tissues. AMPK controls the physiology of multiple organs which are critical to type 2 diabetes, obesity and other metabolic diseases. As such, AMPK regulates both anabolic and catabolic pathways as well as the function and biogenesis of organelles. See text for details.

Advertisement

2. Organization and activation of AMPK

AMPK senses a drop in cellular energy as it is induced by a reduction in glucose availability or other metabolic stresses. The overall consequence of AMPK activation is a change in metabolism; thus, when the AMP/ATP ratio increases AMPK becomes activated in order to rescue the energy balance. As a result of AMPK activation, the cellular metabolism switches from anabolic to catabolic processes. This metabolic shift is accomplished by the AMPK-dependent phosphorylation of multiple targets which are located in different cellular organelles and compartments (see below).

The heterotrimeric enzyme AMPK (Fig. 2; αβγ) contains one catalytic α subunit that is encoded by two genes (α1 and α2). The regulatory β and γ subunits are encoded by two and three genes, respectively (Hardie et al., 2006). The two β subunits (β1, β2) can be myristoylated and phosphorylated, and these modifications may impact the activation and intracellular localization of AMPK (Oakhill et al., 2010; Warden et al., 2001; see below). The γ subunits (γ1, γ2, γ3) bind AMP and ATP in a mutually exclusive fashion, this AMP binding is important to the activation of the enzyme. The subunit composition of AMPK heterotrimers varies in different tissues and can affect the activation of the kinase (Canto & Auwerx, 2010; Cheung et al., 2000; Steinberg & Kemp, 2009; Viollet et al., 2010).

2.1. Control of AMPK activity by phosphorylation and changes in AMPK concentration

The importance of AMPK as a key regulator in cellular metabolism requires a tight control of the enzyme. The rapid regulation of AMPK activity is based on at least three mechanisms that contribute to AMPK activation (Oakhill et al., 2010; Sanders et al., 2007; Shackelford & Shaw, 2009; Steinberg & Kemp, 2009). (a) The most important step for AMPK activation is the phosphorylation of Thr172 of the α subunit which can be modified by the upstream kinases LKB1, CaMKKβ and TAK1 (Fig. 2). Thr172 is phosphorylated when the energy state of the cell is low, i.e. when the AMP/ATP ratio rises. Under these conditions, AMP binding to the regulatory γ subunit promotes the subsequent Thr172 phosphorylation. LKB1 is the major upstream kinase for this event in tissues like skeletal muscle. The effect of AMP binding depends on the type of γ subunit (Cheung et al., 2000). Specifically, AMP-binding to γ2 subunits leads to the largest increase in AMPK activity. By contrast, a relative small change is observed for the γ3 subunit which is mostly synthesized in glycolytic skeletal muscle. Recent data suggest that the β subunits also play a crucial role in AMPK activation. It was proposed that β subunit myristoylation provides a switch that is a prerequisite for Thr172 phosphorylation (Oakhill et al., 2010). (b) Aside from changes in the AMP/ATP ratio, a rise in intracellular Ca2+ concentrations triggers Thr172 phosphorylation. This modification is mediated by CaMKKβ and particularly important in tissues where LKB1 is not the predominant kinase for Thr172. At present, the role of TAK1 in AMPK activation is not fully understood. (c) AMPK activation can be prolonged by preventing the dephosphorylation of Thr172, a process catalyzed by phosphatases PP2A and PP2C (Kim et al., 2009a; Nagata & Hirata, 2010).

Figure 2.

Organization of AMPK and regulation of kinase activity by phosphorylation.AMPK is a heterotrimeric enzyme that is activated by phosphorylation on Thr172 of the α subunit. Several upstream kinases can modify Thr172; they include LKB, CaMKKβ and TAK1. The activity of AMPK can be reduced by different mechanisms. For example, PP2A and PP2C mediate the dephosphorylation of phospho-Thr172. In addition, PKC and Akt phosphorylate Ser485/491 of the α chain which decreases AMPK activity. Furthermore, PKA-dependent modification of Ser173 diminishes AMPK activity.

Aside from the dephosphorylation of phospho-Thr172, a negative regulation of AMPK involves the phosphorylation of Ser485/491 by PKC and possibly Akt, whereas the decline in activity by Ser173 phosphorylation was ascribed to PKA (Djouder et al., 2010). Such modification on Ser173 may help to fine tune lipid metabolism in adipose tissue.

The tissue-specific regulation of AMPK activity is likely achieved by the combined effects of upstream activating kinases, inactivating phosphatases as well as the synthesis and degradation of AMPK subunits. For example, LKB1 is particularly important to activate AMPK in skeletal muscle, whereas CaMKKβ is crucial in the brain (Ronnett et al., 2009). On the other hand, TNFα alters AMPK activation by modulating the synthesis of PP2C (Lu et al., 2010; Steinberg et al., 2006). Aside from the rapid control of AMPK activation by phosphorylation, changes in the expression of subunit genes or the turnover of AMPK subunits can help to fine tune AMPK activity in some tissues (Barry et al., 2010; Fukuyama et al., 2007; Hallows et al., 2006; Qi et al., 2008; Niesler et al., 2007; Steinberg et al., 2003).

2.2. Pharmacological compounds and other factors that alter AMPK activity

Previous work established the essential role of AMPK in the regulation of carbohydrate, protein and lipid metabolism; this made AMPK a key target for the treatment of type 2 diabetes, obesity and metabolic syndrome (Gruzman, Babai & Sasson, 2009; Hardie, 2008b; Steinberg & Kemp, 2009; Viollet et al., 2010; Viollet et al., 2009b). Indeed, in a clinical setting AMPK activity is altered with the anti-diabetic drug metformin and other biguanides. The drug-induced activation of AMPK has important consequences for the patient; among these is the improvement of insulin resistance.

Pharmacological drugs have also been critical to define how AMPK mediates metabolic control (see Table 1). These compounds employ a variety of molecular mechanisms that culminate in AMPK activation (Gruzman et al., 2009; Hawley et al., 2010; Mantovani & Roy, 2011). For example, the kinase can be activated by a rise in the AMP/ATP ratio, generation of an AMP mimetic or increase in intracellular Ca2+ concentrations (Hawley et al., 2010). Metformin impacts several biological processes that ultimately activate AMPK. These include changes in the respiratory chain, increased synthesis of the protein deacetylase SIRT1 (which activates LKB1) and activation of TAK1 (Caton et al., 2010; Hawley et al., 2002; Hawley et al., 2010; Xie et al., 2006). Phenphormin promotes the LKB1-dependent activation of AMPK by inhibiting mitochondrial complex I (Hawley et al., 2010). Resveratrol prevents the acetylation and concomitant inactivation of the upstream kinase LKB1, this compound also inhibits mitochondrial ATP synthase and may increase the concentration of adiponectin (Hawley et al., 2010; Wang et al., 2011). AICAR generates the AMP mimetic 5-amino-4-imidazolecarboxamide ribotide (ZMP) and causes a drop in cellular ATP and ADP, which leads to AMPK activation (Hawley et al., 2010). Aside from drugs that activate AMPK, compound C serves as an ATP-competitive inhibitor of AMPK that has been used widely. All of the compounds discussed here are established pharmacological tools that alter AMPK activation or enzymatic activity; they have been useful for the analysis of AMPK in vitro, in growing cells and in whole animals. The following table summarizes how AMPK activity can be affected in vitro, growing cells, organs or whole organisms.

Compounds, physiological processes and stressMode of action and effect on AMPK
(A) Drugs and other compounds
AICAR (5-aminoimidazole-4-carboxamide-1-b-D-ribofuranoside)Generation of ZMP, which functions as an analog of AMP; activation
Metformin (Biguanide)Reduces mitochondrial ATP production; activation
PhenforminInhibition of respiratory chain, activation
ResveratrolChange in ATP synthase activity; prevents acetylation of LKB1 via modulation of SIRT1; upregulation of adiponectin synthesis and multimerization; activation
Thiazolidinedione derivatives (TZDs, troglitazone; rosiglitazone, pioglitazone)Stimulate the expression and secretion of adiponectin; increase in AMP concentration; activation of PPARγ; AMPK activation
Antimycin AInhibition of respiratory chain; activation
Sodium azideInhibition of respiratory chain; activation
NOInhibition of respiratory chain; activation
OligomycinInhibition of ATP synthase; activation
DinitrophenolUncoupler of electron transfer/ATP synthesis; activation
2-DeoxyglucoseInhibition of glycolysis; activation
ArseniteInhibition of TCA cycle; activation
β-guanadinopropionic acidCreatine analog; increases AMP/ATP ratio; activation
A23187Activation by increase in cytosolic calcium ions
A769662Direct AMPK activator
Compound C (dorsomorphin)Reversible, ATP-competitive inhibitor
(B) Hormones, cytokines, physiological processes and environmental stressors
InsulinInhibition of AMPK activation; mediated by Akt kinase
GhrelinTissue-specific effects; activation in heart and hypothalamus; reduced activity in liver and adipose tissue
AdiponectinActivation by increase in AMP concentration
ResistinTissue-specific effects; reduction of AMPK activity in skeletal muscle
LeptinTissue-specific effects; activates α2 heterotrimers; activation in muscle and fat tissue; reduces activity in hypothalamus
TNFαAcute and chronic effects; acute: activation; chronic: reduction in activity; increase in PP2C
IL-6Increase in AMP/ATP ratio; activation (note that IL-6 can have different effects on insulin sensitivity)
CNTF (Ciliary neurotrophic factor)Tissue-specific effects; activation in muscle; activity reduced in hypothalamus
UCP1, UCP3Uncoupling proteins in mitochondria, change in energy status; activation
Reduction in glucose availabilityChange in energy status; activation
Rise in Ca2+ concentration, osmotic stressCaMKKβ activation
ExerciseSkeletal muscle contraction; activation
Heat shock, oxidative stressEnvironmental stressors; transient activation
Ischemia/hypoxia, reactive oxygen speciesMetabolism/oxidative stress; activation

Table 1.

Modulators of AMPK activity.The data in Table 1 are compiled from several publications that describe the molecular mechanisms and tissue-specific effects on AMPK activity in detail (Caton et al., 2010; Dzamko & Steinberg, 2009; Hawley et al., 2010; Maeda et al., 2001; Nagata & Hirata, 2010; Steinberg et al., 2009; Viollet et al., 2010). It should be noted that although in most cases a correlation between treatment and changes in AMPK activity has been demonstrated, the molecular mechanisms are not always fully understood. For example, hormone or cytokine-dependent changes in AMP/ATP ratios may be secondary to other signaling events, such as changes in cAMP concentrations. For some of the treatments, it has yet to be established whether AMPK is essential for the downstream physiological effect. More recent experiments with knockout cells and animal models will help to fill these gaps (Viollet et al., 2009a).

Advertisement

3. AMPK functions in different tissues and organs

Although AMPK is present in different tissues and organs, the subunit composition varies, and changes in cell physiology can also alter the profile of expressed subunits (Mahlapuu et al., 2004; Pulinilkunnil et al., 2011; Putman et al., 2007; Quentin et al., 2011; Stapleton et al., 1996; Turnley et al., 1999). Of particular importance at the cellular, organ and organismal level is the ability of AMPK to switch from anabolic to catabolic processes when energy supplies are low. AMPK regulates metabolism and other aspects of cell physiology both under normal and disease conditions; studies with different cells or tissues emphasize the significance of AMPK for cellular metabolism and the response to various forms of stress. Thus, AMPK controls several metabolic pathways that are directly relevant to diabetes and other metabolic diseases or syndromes (Steinberg & Kemp, 2009; Viollet et al., 2010; Zhang et al., 2009). However, AMPK not only provides a sensor for nutrient availability, the kinase is also activated by hormonal signals in peripheral tissues and the hypothalamus (Jorda et al., 2010; Ronnett et al., 2009). Notably, this signaling in the central nervous system contributes to the regulation of food uptake. Research with hepatic, skeletal muscle, adipose, pancreatic and kidney cells is particularly important to our understanding of type 2 diabetes as these cell types are crucial to the etiology or pathophysiology of the disease (Fig. 1). In general, the consequences of AMPK activation can be divided into acute and long-term effects (Mantovani & Roy, 2011; Viollet et al., 2010). Whereas the phosphorylation of key enzymes produces a fast downregulation of ATP-consuming metabolic pathways, long-term effects involve changes in the expression of target genes that control metabolism. Since several recently published excellent reviews covered these topics extensively, Table 2 only summarizes the impact of AMPK activation on tissues that are critical to type 2 diabetes.

Tissue or cell typePhysiological processEnzyme or process affected by AMPK
Liveractivation of fatty acid oxidation, inhibition of lipogenesisinhibition of acetyl-CoA carboxylase ACC (Acc1, Acc2)
reduced cholesterol synthesisHMG-CoA reductase
stimulation of fatty acid uptakeCD36 (a fatty acid translocase) moves to the plasma membrane
changes in lipogenesis and glycolysis due to reduced concentration of transcriptional regulators SREBP1 (sterol response element binding protein-1) and ChREBP (carbohydrate response-element binding protein)inhibits ChREBP by phosphorylation, reduces the transcription of genes encoding SREBP1 and ChREBP
increase in mitochondrial biogenesisincreased expression of PGC1α and other genes required for mitochondrial biogenesis
glycogen synthesis reducedinhibition of glycogen synthase
inhibition of gluconeogenesis and hepatic glucose productionchanges in the activity, concentration or localization of key enzymes or transcriptional regulators; (phosphoenol pyruvate carboxy kinase, HNF4; TORC2, p300)
Skeletal musclestimulation of glucose uptake; fusion of GLUT4 (glucose transporter) containing vesicles with plasma membranephosphorylation of AS160 may promote trafficking of vesicles; increased transcription of GLUT4 gene by phosphorylation of HDAC5
increase in mitochondrial biogenesisincreased expression of PGC1α and other genes required for mitochondrial biogenesis
increased fatty acid uptake and oxidationinhibition of ACC
reduction in protein synthesisinhibition of mTOR pathway via modification of mTOR, TSC2 and eEF2 kinase
control of glycogen metabolisminactivation of glycogen synthase
Adipose tissueincrease in fatty acid oxidationinactivation of ACC
inhibition of lipolysisphosphorylation of HSL, reduced association of HSL with lipid droplets
Pancreasinhibition of glucose-induced insulin secretion in β cellsreduced trafficking of vesicles containing insulin
inhibition of transcription of the preproinsulin gene in β cells;
stimulation of glucagon secretion in α cells
molecular mechanisms not fully understood
Heartstimulation of glucose uptake by translocation of GLUT4 to the plasma membranefusion of GLUT4 containing vesicles with the plasma membrane
stimulation of glycolysisactivation of 6-phosphofructo-2-kinase → enhances production of fructose 2,6-bisphosphate → stimulates 6-phosphofructo-1-kinase
increase in fatty acid oxidationinactivation of ACC
control of glycogen metabolismcontributions of AMPK activity not completely understood at the molecular level
Kidneyameliorates changes linked to diabetic nephropathyinhibition of mTOR, inhibition of CFTR and other ion channels
Brainfood intake; multiple pathways affected in the hypothalamus; adiponectin, leptin, insulin and ghrelin control AMPKcontrol of neuropeptide synthesis

Table 2.

AMPK-dependent modulation of cell physiology. Some of the tissue-specific reactions regulated by AMPK and relevant to type 2 diabetes are listed. A comprehensive description of these processes can be found in several recent reviews (Hardie, 2008a; Ix & Sharma, 2010; Lieberthal & Levine, 2009; Steinberg & Kemp, 2009; Viollet et al., 2009a; Viollet et al., 2010) and original publications (da Silva Xavier et al., 2003; Leclerc et al., 2011; Takiar et al., 2011; van Oort et al., 2009).

Advertisement

4. AMPK modulates targets in different subcellular organelles and compartments

4.1. Subcellular distribution of AMPK substrates

The combination and integration of different subcellular events regulated by AMPK enables cells, tissues and organs to coordinate different metabolic pathways in order to achieve and maintain the proper energy balance of the whole organism. Fig. 3 depicts established AMPK substrates according to their presence in different subcellular compartments. Table 3 expands this information and specifies how the AMPK-dependent phosphorylation of individual substrates alters their functions.

It is obvious from Fig. 3 that AMPK phosphorylates a large number of proteins that are associated with distinct organelles or subcellular compartments. Cytoplasmic and mitochondrial substrates of the kinase include enzymes that are involved in fat, protein, glucose and glycogen metabolism. Kinase targets in the plasma membrane consist of ion channels, carriers and receptors, whereas other substrates are linked to the function or trafficking of intracellular membranes. This includes the transport of vesicles containing the glucose transporter GLUT4, because GLUT4 translocation to the plasma membrane is a pre-requisite for efficient glucose uptake in skeletal muscle and other tissues. AS160 and TBC1D1 likely play a role in these processes (Table 3). In the nucleus, the AMPK-mediated modification of transcription factors, transcriptional regulators and a subunit of RNA-polymerase I control the expression of genes that are implicated in specific anabolic and catabolic reactions. The phosphorylation of several of these targets is also critical to the biogenesis of mitochondria and the assembly of ribosomes.

Figure 3.

AMPK affects functions in various cellular compartments and organelles. Examples are shown for the proteins that AMPK modifies in distinct sub-cellular locations. Some of the substrates are present in multiple compartments. PM, plasma membrane. See text and Table 3 for details.

Given the diverse types of AMPK substrates and their presence in different cellular locations, it is helpful to recapitulate their functions (Table 3a). This knowledge is a prerequisite to understand how the dynamic association and action of AMPK in different compartments will impact downstream events.

Substrates that have been established for AMPK heterotrimers that contain the α1 or α2 subunit are shown. For different AMPK substrates the function, effect of AMPK-dependent phosphorylation and the major subcellular localization are depicted. For some substrates, there are cell-type specific differences, and the effect of AMPK-dependent phosphorylation may not be fully understood or controversial. The list of AMPK substrates was compiled from PhosphoSitePlus (phosphosite.org).

Substrates for AMPKα1FunctionEffect of phos-phorylationPrimary intracellular localizationRefe-rences
Acc1;
Subunit of acetyl-CoA carboxylase, ACC
Carboxylates acetyl-CoA, thereby generating malonyl-CoA; this step is rate-limiting for FA biosynthesis.inhibitioncytoplasmSun et al., 2006
Acc2;
Subunit of acetyl-CoA carboxylase, ACC
Carboxylates acetyl-CoA, thereby generating malonyl-CoA; this step is rate-limiting for FA biosynthesis.inhibitionmitochondria (outer mitochondrial membrane)Reihill et al., 2007
AMPKα1Catalytic subunit of AMPKpotential autoregulationcytoplasm, nucleusStein et al., 2000
AMPKβ1Regulatory subunit of AMPKenzymatic activity, localizationcytoplasm, nucleusWarden et al., 2001
CFTRCystic fibrosis transmembrane conductance regulator, chloride channelinhibits PKA-dependent stimulation of CFTRplasma membrane, ER, cytoplasmic vesicle; early endosomeKing et al., 2009; Kong-suphol et al., 2009; Takiar et al., 2011
ChREBPTranscription factor; repressorinactivation of DNA bindingnucleus, cytoplasmKawagu-chi et al., 2002
CK1-ε (CK1 epsilon)Ser/thr kinase; member of the casein kinase 1 (CK1) family; phosphorylates mPer2increase of CK1-ε activity; control of circadian clockcytoplasm, nucleus, cell junction, centrosomeUm et al., 2007
CRY1 (crypto-chrome)DNA photolyase, regulates circadian rhythmphosphorylation leads to destabilizationnucleus, mitochondria, cytoplasmLamia et al., 2009
CRTC-1CREB-regulated transcriptional coactivatorphosphorylation causes cytoplasmic retentionnucleus, cytoplasmMair et al., 2011
eEF2K
(eEF2 kinase)
Protein kinase; eEF2K modifies and thereby inactivates eEF2activation of kinase activitycytoplasmBrown et al., 2004
eNOSEndothelial nitric oxide synthase (constitutive), EC 1.14.13.39; regulation of cytoskeletal
reorganization
activation, promotes deacetylation by SIRT1plasma membrane, GolgiChen et al., 2010; Sun et al., 2006
GABAB-R1GABAB receptor subunit; G-protein coupled receptorincrease in receptor function, promotes functional coupling to K+ channelsplasma membrane, ER, dendrite, axon, postsynaptic membraneKura-moto et al., 2007
GABAB-R2GABAB receptor subunit; G-protein coupled receptorincrease in receptor function, promotes functional coupling to K+ channelsplasma membrane, ER, dendrite, axon, postsynaptic membraneKura-moto et al., 2007
GBF1; Golgi-specific brefeldin A resistance factorGuanine nucleotide exchanger for ARF5; mediates ARF5 activation; Golgi to ER traffickingtriggers disassembly of Golgi apparatusGolgi membraneMiya-moto et al., 2008
GFATglutamine-fructose-6-phosphate transaminase 1; EC 2.6.1.16; regulates glucoseflux to hexosamine pathway1.4 fold increase in enzymatic activitycytoplasmLi et al., 2007
GYS1Glycogen synthase; EC 2.4.1.11inhibits enzymatic activitycytoplasmSkurat, et al., 2000
H2BHistone H2Bactivates stress-induced transcriptionnucleusBungard et al., 2010
HDAC5Histone deacetylase 5; EC3.5.1.98; transcriptional regulator, cell cycle progression, developmentreduced binding to GLUT4 promoter → enhanced GLUT4 expressionnucleus
(cytoplasm)
McGee et al., 2008
HNF4 alphaHepatocyte nuclear factor 4 alpha; transcription factor, control of gene expression in hepatocytesreduced DNA-bindingnucleusHong et al., 2003
HSLHormone sensitive lipase; EC 3.1.1.79; involved in triglyceride lipolysisinhibition; change in the association with lipid dropletscytoplasm,
lipid droplets
Garton et al. 1989; ; Watt et al., 2006
IRS1;
adaptor protein
Insulin receptor substrate 1, adaptor proteinmodulation of PI3 kinase signalingcytoplasmJakobsen et al., 2001; Tzatsos & Tsich-lis, 2007
KCNMA1 iso4;
α subunit of BKCa channel
Potassium channel, activated in response to membrane depolarization, oxygen sensing in carotid bodyinhibition of potassium currentsintegral membrane protein, plasma membrane, axonRoss et al., 2011
Kir6.2
(KCNJ11)
Potassium channel, voltage dependent, inward rectifyingmight play a role in insulin secretionplasma membraneChang et al., 2009
KxlinkKinesin 2; kinesin light chain 1, motor proteinbiological role of modification not knowncytoplasm, microtubulesMcDo-nald et al., 2009, 2010
KPNA2, importin-α1Adaptor for classical nuclear importinterferes with nuclear import of HuRcytoplasm, nucleus, nuclear envelopeWang et al., 2004
mTORSer/thr kinase; EC 2.7.11.1; catalytic subunit of mTORC1 and mTORC2links nutrient supply to translationcytoplasmCheng et al., 2004
NKCC2 (SLC12A1)Electroneutral transporter; reabsorption of Na+ and Cl¯; controls cell volumeregulation of transporter activityplasma membraneFraser et al., 2007
p27Kip1 (CDKN4)Cyclin-dependent kinase inhibitor 1B, controls cell cycle progression at G1stabilization of p27; linked to autophagynucleusLiang et al., 2007
p300Protein (histone) acetyltransferase; EC2.3.1.48; transcriptional co-activatorinhibits interaction with nuclear receptors, such as PPARγnucleusYang et al., 2001
p53Tumor suppressor, transcription factor, cell cycle arrest, DNA repair, apoptosisstabilization of p300-p53 interaction; controls cell cycle progressionnucleusDornan & Hupp, 2001
PFKFB26-Phosphofructo-2-kinase/ fructose-2,6-bisphosphatase 2; EC 2.7.1.105 or EC 3.1.3.46; glycolysisactivation;
stimulation of glycolysis
cytoplasmMarsin et al., 2000
PFKFB3;
iPFK2
similar to PFKFB2; inducible in monocytesactivationcytoplasmMarsin et al., 2002
PPP1R3C
(R5/PTG)
Regulatory subunit of protein phosphatase PP1; controls PP1 activity; targets PP1 to glycogen; stimulation of glycogen synthasepromotes ubiquitination and thereby degradationglycogen granulesVernia et al., 2009
PPP2R5C
(B56 γ)
Regulatory subunit of protein phosphatase PP2A; possible role in the regulation and targeting of PP2Aincreases
PP2A activity
nucleus, chromosomesKim et al., 2009b
Raf1Ser/thr kinase; EC 2.7.11.1; component of Ras→Raf→MEK1/2→ERK1/2 signaling pathwaycytoplasm, plasma membraneSprenkle et al., 1997
RaptorRegulation of mTORC1 (mammalian target of rapamycin complex 1); functions as scaffoldinhibition of mTORC1; cell cycle arrestcytoplasmGwinn et al., 2008
RbRetinoblastoma protein; regulates cell cycle progression at G1; functions as transcriptional co-regulator; tumor suppressorcontrol of brain developmentnucleusDasgupta & Milbrandt, 2009
smMLCKSmooth muscle myosin light chain kinase; ser/thr protein kinase; EC2.7.11.18reduces activity of smMLCKcytoplasmHorman et al., 2008
TBC1D1GTPase activating protein Rab family members; regulates glucose transportinduces binding to 14-3-3 proteinsERChen et al., 2008
TIF-IATranscription initiation factor for RNA-pol Ireduced rDNA transcriptionnucleolusHoppe et al., 2009
TORC2 (CRTC2)Transducer of regulated CREB protein 2; transcriptional regulatorphosphorylation causes cytoplasmic retentionnucleus, cytoplasmKoo et al., 2005
TSC2 (tuberin)Generates heterodimer with hamartin (TSC1); TSC1/TSC2 functions as GTPase activator of Rhebenhances TSC2 activity → inhibition of mTORcytoplasmInoki, et al., 2003
ULK1Ser/thr kinase; EC 2.7.11.1; binds to mTORC1 via raptor, binding controlled by nutrient supplycontrol of autophagycytoplasmEgan et al., 2011
VASPVasodilator-stimulated phosphoprotein; actin regulatorimpairs endothelial actin assemblycytoplasm, cytoskeletonBlume et al., 2007
ZNF692
(AREBP)
Transcriptional regulator, contains zinc fingerreduction in DNA bindingnucleusInoue & Yamau-chi, 2006

Table 3.

Substrates for AMPKα2FunctionEffect of phos-phorylationPrimary intracellular localizationRefe-rences
ACC1See above description of AMPKα1 targetscytoplasm
AS160 (TBC1D4; Akt substrate of 160k)GTPase activating protein for Rab; implicated in GLUT4 exocytosis in skeletal musclenot fully understood;
may regulate glucose uptake
cytoplasmEguez et al., 2005; Treebak et al., 2010
HAS2Hyaluronic acid synthase 2; EC 2.4.1.212; integral membrane proteininhibition of enzymatic activityplasma membraneVigetti et al., 2011
HDAC5See above description of AMPKα1 targetsnucleus
p53See above description of AMPKα1 targetsnucleus
PGC1αPPAR γ coactivator-1; transcriptional co-activator; association with PPARγ; binds to CREB and nuclear respiratory factors; controls mitochondrial biogenesisphosphorylation alters activity as transcriptional
regulator
nucleusJager et al., 2007
PLD1Phospholipase D1 phophatidylcholine specific; EC 3.1.4.4; linked to Ras signaling; involved in membrane traffickingactivation of enzymatic activityGolgi, ERKim et al., 2010

Table 3a.

Substrates of AMPK heterotrimers containing the α1 or α2 subunit

4.2. Subcellular distribution and trafficking of AMPK

AMPK is associated with different organelles and subcellular compartments; however, little is known about the dynamic nature of this distribution. Analyses of other signaling pathways have demonstrated that the subcellular localization of kinases is critical for the proper response to extra- and intracellular stimuli, and it is likely that the same scenario applies to AMPK. We will therefore briefly review what is currently known about the subcellular localization and trafficking of AMPK.

AMPK is found in the nucleus and cytoplasm which reflects functions for the enzyme in both locations (Kodiha et al., 2007; Witczak et al., 2008). Although the kinase is associated with multiple compartments, α1 and α2 subunits differ in their nucleocytoplasmic distribution. Under normal growth conditions, α1 is predominantly in the cytoplasm, whereas α2 locates to both the nucleus and cytoplasm (Salt et al., 1998). It was further shown that the nuclear localization sequence (NLS) present in the α2 subunit is required for AMPK nuclear translocation (Suzuki et al., 2007), suggesting that the catalytic α subunit is essential for the proper intracellular localization of the holoenzyme. Our current model of AMPK trafficking proposes that the kinase shuttles between the nucleus and the cytoplasm. Data from our group and others support this idea, as they demonstrated that the nuclear carrier Crm1 is essential for AMPK export from the nucleus to the cytoplasm (Kazgan et al., 2010; Kodiha et al., 2007).

How the β subunit impacts the proper targeting of the holoenzyme is at present not entirely clear. The β1 subunit can be modified posttranslationally, both by phosphorylation and myristoylation, and these modifications were linked to the subcellular targeting of the β1 subunit (Suzuki et al., 2007; Warden et al., 2001). It was proposed that AMPK concentrates in the cytoplasm when the heterotrimeric enzyme contains the β1 subunit (Suzuki et al., 2007). However, this model is difficult to reconcile with the fact that both β1 and β2 subunits can be detected in the nucleus (Kodiha et al., 2007). Furthermore, recent studies suggest that the myristoylation of β1 and β2 subunit is particularly important for AMPK activation, as AMP-dependent myristoylation provides a switch that triggers Thr172 phosphorylation (Oakhill et al., 2010). Although the contribution of β subunits to nuclear and membrane targeting of the holoenzyme is not completely understood at this point, the importance of the β subunit for glycogen binding is well established (Polekhina et al., 2003).

In contrast to the α and β subunits, little is known about the trafficking of AMPK γ subunits. In Drosophila, the single γ subunit migrates into the nucleus of the fat body with the onset of autophagy during normal development, and a potential NLS was detected in this subunit (Lippai et al., 2008). It was speculated that this nuclear accumulation contributes to the expression of genes that are necessary for autophagy.

Several studies support the model that the intracellular distribution of AMPK in human and other cells is dynamic. This is particularly important in the context of disease, because the distribution of AMPK can be modulated by physiological and environmental stimuli. For example, the α2 subunit translocates to the cell nucleus upon exercise or environmental stress (Kodiha et al., 2007; McGee et al., 2003), indicating that the adaptation of skeletal muscle during exercise or metabolic stress is at least in part mediated by the subcellular relocation of AMPK. Examples of the relocation of AMPK α subunits in human cells exposed to oxidative stress or depleted for energy are shown in Fig. 4 (Kodiha et al., 2007).

The relocation of AMPK subunits in response to physiological changes is not restricted to the α subunits; our previous experiments demonstrated that AMPK β subunits accumulate in the nucleus as well when cells are exposed to oxidative and other forms of stress (Kodiha et al., 2007). Moreover, AMPK localization could be regulated by the circadian rhythm. Specifically, changes in the expression of AMPK subunits may depend on the circadian rhythm; this change in expression will then alter the intracellular distribution of AMPK (Lamia et al., 2009). Since these studies were carried out with mice, it has yet to be shown whether the same applies to humans.

Figure 4.

AMPKα concentrates in nuclei when cells are exposed to oxidative stress or depleted for energy. HeLa cells were treated with diethyl maleate to induce oxidative stress or with a combination of sodium azide and 2-deoxyglucose for energy depletion. The distribution of AMPKα1 and α2 subunits was examined by indirect immunofluorescence; DNA was stained with DAPI (Kodiha et al., 2007). Note that the α-subunits are more concentrated in nuclei of stressed cells. Several nuclei are marked with arrowheads. Size bar is 20 μm.

Studies from several laboratories, including our group, defined the signals and mechanisms that determine the trafficking and intracellular distribution of AMPK. Our work also suggested crosstalk between other signaling cascades and the localized action of AMPK (Kodiha et al., 2007 and unpublished). Ultimately, such crosstalk will add to the complexity of downstream events that are modulated by AMPK. Taken together, previous research suggests that AMPK subunits move between different subcellular locations, and it can be expected that the compartment-specific actions of the kinase are linked to the physiological response of cells and tissues.

4.3. How does the compartment-specific action of AMPK impact cellular functions that are relevant to type 2 diabetes?

Type 2 diabetes is associated with the increased risk of a growing number of diseases and pathologies. This is exemplified by renal nephropathy, myocardial disease, stroke, Alzheimer’s and Parkinson’s disease (Almdal et al., 2004; Biessels et al., 2006; Burdo et al., 2009; Hallows et al., 2010; Hu et al., 2007; Maher & Schubert, 2009; Schernhammer et al., 2011). Several drugs are currently used in the clinical setting to activate AMPK in patients suffering from type 2 diabetes or obesity. However, it should be kept in mind that AMPK activation can be beneficial as well as harmful in the ischemic heart, and AMPK activation may be linked to neurodegeneration (Lopaschuk, 2008; Spasic, Callaerts & Norga, 2009; Thornton et al., 2011; Vingtdeux et al., 2011). Thus, activation of AMPK throughout the whole organism or the entire cell of a particular tissue may not always be advantageous. As an alternative approach, we put forward the concept of a compartment-specific modulation of AMPK action. Since AMPK activation can be damaging in the context of some of the complications associated with type 2 diabetes, our approach applies both to the localized activation as well as inhibition of the kinase. We believe that the confined action of AMPK will provide a better therapeutic approach in the future that could reduce the side-effects of AMPK modulators. The simplified model in Fig. 5 summarizes the possible changes of cellular functions that will be induced by targeting AMPK in different subcellular compartments.

Figure 5.

Localized modulations of AMPK activity. The possible changes induced by the compartment-specific alteration of AMPK activity are depicted. Note that there are cell-type dependent differences for the processes regulated by AMPK.

In the past few years, significant progress has been made with the identification of AMPK substrates and their links to human disease. As shown in Fig. 3 and Table 3, AMPK modifies targets in different subcellular compartments or organelles. We propose that the modification of these substrates relies on (a) the amount of active AMPK and (b) the intracellular distribution of AMPK. The combination of AMPK activation and subcellular localization will then determine the level of phosphorylation of its substrates and the subsequent changes in cell physiology. Such changes will affect both the rapid response to specific stimuli as well as the long-term modification of metabolism and other processes. In the following, we focus on some of the processes that are controlled by AMPK and linked to type 2 diabetes or the complications associated with the disease.

4.3.1. AMPK targets the protein synthesis apparatus in the cytoplasm

Several of the cytoplasmic substrates of AMPK are essential to promote a fast cellular response to changes in nutrient supply. For example, AMPK phosphorylates cytoplasmic targets, such as eEF2 kinase and mTOR, which regulate protein translation. AMPK-dependent modification of these substrates results in downregulation of protein synthesis. Under some conditions, it could be desirable to preferentially modulate these processes that are associated with cytoplasmic AMPK targets. A possible example of such a scenario is the diabetic kidney (Cammisotto et al., 2008; Hallows et al., 2010; Lee et al., 2007; McMahon et al., 2009). Diabetes-induced renal hypertrophy correlates with diminished AMPK activity and, at the same time, with increased protein synthesis under high glucose conditions (Hallows et al., 2010). AICAR and metformin reduce protein synthesis triggered by high glucose (Lee et al., 2007), but these compounds also produce effects that are unrelated to AMPK activation (Mantovani & Roy, 2011). Thus, stimulating AMPK in the cytoplasm could provide a more focused approach to reduce damage in the diabetic kidney.

4.3.2. AMPK targets associated with the plasma membrane and vesicular trafficking

Several channels and transporters in the plasma membrane are phosphorylated by AMPK and control the secretion of insulin, hyperpolarization of β-cells under low glucose concentration and the response to hypoxia (Beall et al., 2010; Chang et al., 2009; Düfer et al., 2010; Evans et al., 2009; Hallows, 2005; Zheng et al., 2008). Although details of the molecular mechanisms are not always clear and in part controversial, altering the AMPK activity at the plasma membrane has the potential to modify β-cell function. The same principle could also apply to the heart and kidney, where several integral proteins of the plasma membrane are modified by AMPK.

4.3.3. AMPK substrates in the nucleus

In the nucleus, AMPK directly regulates the transcription of genes that control metabolism as well as the biogenesis of mitochondria and ribosomes. As such, AMPK modifies HNF4α, HDAC5, p300, histone H2B, the tumor suppressor p53, PGC1α, and TIF-1A (see Fig. 3 and Table 3). AMPK-dependent phosphorylation of these targets is critical to alter the transcriptional profile, which in turn is necessary to adjust metabolic activities in skeletal muscle, heart, liver and adipose tissues in response to changes in glucose availability. It is reasonable to assume that the modification of transcription factors and transcriptional regulators will rely to a large extent on AMPK in the nucleus. This model is supported by a recent study that shows AMPK to move along genes together with the transcriptional machinery (Bungard et al., 2010). Activation of AMPK in the nucleus could enhance the effects of AMPK on the transcription of several target genes. One of the possible benefits of the activation of nuclear AMPK will be the increase in mitochondrial biogenesis. In the long-term, this could help to stimulate the oxidation of fatty acids and limit the lipotoxicity that is linked to type 2 diabetes (Schrauwen & Hesselink, 2004).

4.3.4. AMPK targets associated with mitochondria

Acc2 is associated with mitochondria and important for the synthesis of malonyl-CoA, an intermediate of fatty acid biosynthesis. AMPK phosphorylates and thereby inactivates Acc, which leads to a reduction in malonyl-CoA concentration. As a consequence, de novo fatty acid synthesis is reduced and fatty acid oxidation is upregulated. It is conceivable that the localized Acc2 inhibition by AMPK could stimulate CPT-1 (carnitine palmitoyltransferase-1) dependent transport of fatty acids into mitochondria for subsequent degradation. This in turn could reduce the load of peroxidation products of fatty acids in the cytoplasm and the subsequent damage to mitochondria (Schrauwen & Hesselink, 2004).

Advertisement

5. Development of drugs that alter the compartment-specific activity of AMPK

In order to modulate AMPK activity in a fashion that is more localized as compared to the currently used drugs, a number of questions will have to be addressed. This includes the ability to regulate AMPK (a) in different organs or tissues and (b) in specific subcellular locations. Oral administration of metformin is believed to preferentially alter liver metabolism, whereas TZDs and their derivatives affect adipose tissue, skeletal muscle and probably β-cells (Gruzman et al., 2009). One possibility to enhance the tissue-specific action of AMPK will rely on the development of drugs that directly bind to AMPK; indeed such compounds have been described (Hawley et al., 2010). Since tissue-specific differences in AMPK subunits have been established, developing compounds that preferentially interact with individual subunits or specific subunit combinations of heterotrimers could provide a means to increase the specificity of AMPK action. For example, the γ3 subunit is predominantly synthesized in glycolytic skeletal muscle and could therefore serve as a target to alter AMPK in this tissue.

Taking into account differences in AMPK subunits could further be exploited to regulate the kinase in different subcellular locations (see section 4.2). Thus, it is believed that the α2 subunit is more concentrated in the nucleus as compared to the α1 subunit; this difference could help to activate mainly nuclear or cytoplasmic pools of the kinase. This line of reasoning could be expanded to the posttranslational modifications of the β subunit, as phosphorylation and myristoylation of the β subunits are implicated in the subcellular distribution of the kinase.

In addition to taking advantage of the differences in AMPK heterotrimers, developing AMPK modulators that accumulate in distinct subcellular compartments will be useful. This strategy could be based on the generation of combimolecules with multiple properties (Rachid et al., 2007). Combimolecules that combine DNA-binding with AMPK activation could enhance the modification of nuclear substrates and thereby alter the gene expression profile. On the other hand, such combimolecules could exploit the differences in lipid composition of intracellular membranes to control the AMPK-dependent phosphorylation of mitochondrial Acc2 or channels residing in the plasma membrane.

Advertisement

6. Conclusions

AMPK and its substrates are critical to the etiology and pathology of type 2 diabetes and other metabolic diseases. Several of the current therapeutic regimens for type 2 diabetes alter AMPK activity, either by affecting the cellular energy status or the concentration of AMPK modulators. More recent studies led to the identification of compounds that directly bind AMPK and change its enzyme activity. We propose that the future design of drugs that takes into account the dynamic subcellular distribution of the kinase and its substrates will help to regulate AMPK not only in different tissues and organs, but also at the subcellular level. In the long term this approach will help to fine-tune AMPK action and the downstream events that rely on the phosphorylation of its targets.

References

  1. 1. AlmdalT.ScharlingH.JensenJ. S.VestergaardH.2004The Independent Effect of Type 2 Diabetes Mellitus on Ischemic Heart Disease, Stroke, and Death: A Population-Based Study of 13 000 Men and Women With 20 Years of Follow-up.Arch Intern Med 16414221426
  2. 2. AmatoS.LiuX.ZhengB.CantleyL.RakicP.ManH. Y.2011AMP-Activated Protein Kinase Regulates Neuronal Polarization by Interfering with PI 3-Kinase LocalizationScience332247251
  3. 3. BarryS. P.LawrenceK. M.Mc CormickJ.et al.2010New targets of urocortin-mediated cardioprotectionJ Mol Endocrinol 456985
  4. 4. BeallC.PiipariK.Al-QassabH.SmithM. A.ParkerN.CarlingD.ViolletB.WithersD. J.AshfordM. L. J.2010Loss of AMP-activated protein kinase alpha2-subunit in mouse beta-cells impairs glucose-stimulated insulin secretion and inhibits their sensitivity to hypoglycaemia.Biochem J 429323333
  5. 5. BiesselsG. J.StaekenborgS.BrunnerE.BrayneC.ScheltensP.2006Risk of dementia in diabetes mellitus: a systematic reviewLancet Neurol 56474
  6. 6. BlumeC.BenzP. M.WalterU.HaJ.KempB. E.RenneT.2007AMP-activated Protein Kinase Impairs Endothelial Actin Cytoskeleton Assembly by Phosphory-lating Vasodilator-stimulated Phosphoprotein. J Biol Chem 28246014612
  7. 7. BrowneG. J.FinnS. G.ProudC. G.2004Stimulation of the AMP-activated Protein Kinase Leads to Activation of Eukaryotic Elongation Factor 2 Kinase and to Its Phosphorylation at a Novel Site, Serine 398.J Biol Chem 2791222012231
  8. 8. BungardD.FuerthB. J.ZengP. Y.FaubertB.MaasN. L.ViolletB.CarlingD.ThompsonC. B.JonesR. G.BergerS. L.2010Signaling kinase AMPK activates stress-promoted transcription via histone H2B phosphorylationScience32912011205
  9. 9. BurdoJ. R.ChenQ.CalcuttN. A.SchubertD.2009The pathological interaction between diabetes and presymptomatic Alzheimer’s diseaseNeurobiol Aging 3019101917
  10. 10. CammisottoP. G.BendayanM.CammisottoP. G.BendayanM.2008Adiponectin stimulates phosphorylation of AMP-activated protein kinase alpha in renal glomeruli.J Mol Histol 39579584
  11. 11. CantoC.AuwerxJ.2010AMP-activated protein kinase and its downstream transcriptional pathwaysCMLS 6734073423
  12. 12. CatonP. W.NayuniN. K.KieswichJ.KhanN. Q.YaqoobM. M.CorderR.2010Metformin suppresses hepatic gluconeogenesis through induction of SIRT1 and GCN5J Endocrinol 20597106
  13. 13. ChangT. J.ChenW. P.YangC.LuP. H.LiangY. C.SuM. J.LeeS. C.ChuangL. M.2009Serine-385 phosphorylation of inwardly rectifying K+ channel subunit (Kir6.2) by AMP-dependent protein kinase plays a key role in rosiglitazone-induced closure of the K(ATP) channel and insulin secretion in rats. Diabetologia 5211121121
  14. 14. ChenS.MurphyJ.TothR.CampbellD. G.MorriceN. A.MackintoshC.2008Complementary regulation of TBC1D1 and AS160 by growth factors, insulin and AMPK activatorsBiochem J 409449459
  15. 15. ChenZ.PengI. C.CuiX.LiY. S.ChienS.ShyyJ. Y.2010Shear stress, SIRT1, and vascular homeostasis. Proc Natl Acad Sci USA 1071026810273
  16. 16. ChengS. W. Y.FryerL. G. D.CarlingD.ShepherdP. R.2004Thr2446 Is a Novel Mammalian Target of Rapamycin (mTOR) Phosphorylation Site Regulated by Nutrient Status.J Biol Chem 2791571915722
  17. 17. CheungP. C.SaltI. P.DaviesS. P.HardieD. G.CarlingD.2000Characterization of AMP-activated protein kinase gamma-subunit isoforms and their role in AMP binding.Biochem J 346659669
  18. 18. DaSilva.XavierG.LeclercI.VaradiA.TsuboiT.MouleS. K.RutterG. A.2003Role for AMP-activated protein kinase in glucose-stimulated insulin secretion and preproinsulin gene expression.Biochem J 371761774
  19. 19. DasguptaB.MilbrandtJ.2009AMP-Activated Protein Kinase Phosphorylates Retinoblastoma Protein to Control Mammalian Brain DevelopmentDevelopm Cell 16256270
  20. 20. DjouderN.TuerkR. D.SuterM.SalvioniP.ThaliR. F.ScholzR.VaahtomeriK.AuchliY.RechsteinerH.BrunisholzR. A.ViolletB.MakelaT. P.WallimannT.NeumannD.KrekW.2010PKA phosphorylates and inactivates AMPKα to promote efficient lipolysis. EMBO J 29469481
  21. 21. DornanD.HuppT. R.2001Inhibition of 53 transcription by BOX-I phospho-peptide mimetics that bind to p300.EMBO Rep, 139-144.
  22. 22. DüferM.NoackK.Krippeit-DrewsP.DrewsG.2010Activation of the AMP-activated protein kinase enhances glucose-stimulated insulin secretion in mouse beta-cellsIslets2156163
  23. 23. DzamkoN. L.SteinbergG. R.2009AMPK-dependent hormonal regulation of whole-body energy metabolism. Acta Physiologica196115127
  24. 24. EganD. F.ShackelfordD. B.MihaylovaM. M.et al.2011Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagyScience331456461
  25. 25. EguezL.LeeA.ChavezJ. A.MiineaC. P.KaneS.LienhardG. E.Mc GrawT. E.2005Full intracellular retention of GLUT4 requires AS160 Rab GTPase activating protein.Cell Metabolism2263272
  26. 26. EvansA. M.HardieD. G.PeersC.et al.2009Ion Channel Regulation by AMPK. Annals New York Acad Sci 117789100
  27. 27. FogartyS.HardieD. G.2010Development of protein kinase activators: AMPK as a target in metabolic disorders and cancer.Biochim Biophys Acta 1804581591
  28. 28. FraserS. A.GimenezI.CookN.JenningsI.KaterelosM.KatsisF.LevidiotisV.KempB. E.PowerD. A.2007Regulation of the renal-specific Na+-K+-2Cl- co-transporter NKCC2 by AMP-activated protein kinase (AMPK). Biochem J 4058593
  29. 29. FukuyamaY.OhtaK.OkoshiR.SueharaM.KizakiH.NakagawaK.2007Hypoxia Induces Expression and Activation of AMPK in Rat Dental Pulp Cells.J Dental Res 86903907
  30. 30. GartonA. J.CampbellD. G.CarlingD.HardieD. G.ColbranR. J.YeamanS. J.1989Phosphorylation of bovine hormone-sensitive lipase by the AMP-activated protein kinase. Eur J Biochem 179249254
  31. 31. GruzmanA.BabaiG.SassonS.2009Adenosine Monophosphate-Activated Protein Kinase (AMPK) as a New Target for Antidiabetic Drugs: A Review on Metabolic, Pharmacological and Chemical ConsiderationsRev Diabetic Studies 61336
  32. 32. GwinnD. M.ShackelfordD. B.EganD. F.MihaylovaM. M.MeryA.VasquezD. S.TurkB. E.ShawR. J.2008AMPK Phosphorylation of Raptor Mediates a Metabolic CheckpointMol Cell 30214226
  33. 33. HallowsK. R.2005Emerging role of AMP-activated protein kinase in coupling membrane transport to cellular metabolism.Current Opinion Nephrol Hypertens 14464471
  34. 34. HallowsK. R.FitchA. C.RichardsonC. A.ReynoldsP. R.ClancyJ. P.DagherP. C.WittersL. A.KollsJ. K.PilewskiJ. M.2006Up-regulation of AMP-activated Kinase by Dysfunctional Cystic Fibrosis Transmembrane Conductance Regulator in Cystic Fibrosis Airway Epithelial Cells Mitigates Excessive Inflammation.J Biol Chem 28142314241
  35. 35. HallowsK. R.MountP. F.Pastor-SolerN. M.PowerD. A.2010Role of the energy sensor AMP-activated protein kinase in renal physiology and diseaseAmerican Journal of PhysiologyRenal Physiology 298, F1067F1077.
  36. 36. HardieD. G.2008aAMPK: a key regulator of energy balance in the single cell and the whole organismInt J Obesity 32 S4, S712
  37. 37. HardieD. G.2008bRole of AMP-activated protein kinase in the metabolic syndrome and in heart diseaseFEBS Lett 5828189
  38. 38. HardieD. G.HawleyS. A.ScottJ. W.2006AMP-activated protein kinase- development of the energy sensor concept. J Physiol 574715
  39. 39. HawleyS. A.GadallaA. E.OlsenG. S.HardieD. G.2002The Antidiabetic Drug Metformin Activates the AMP-Activated Protein Kinase Cascade via an Adenine Nucleotide-Independent Mechanism.Diabetes5124202425
  40. 40. HawleyS. A.RossF. A.ChevtzoffC.et al.2010Use of cells expressing gamma subunit variants to identify diverse mechanisms of AMPK activation.Cell Metabolism11554565
  41. 41. HongY. H.VaranasiU. S.YangW.LeffT.2003AMP-activated Protein Kinase Regulates HNF4α Transcriptional Activity by Inhibiting Dimer Formation and Decreasing Protein Stability. J Biol Chem 2782749527501
  42. 42. HoppeS.BierhoffH.CadoI.WeberA.TiebeM.GrummtI.VoitR.2009AMP-activated protein kinase adapts rRNA synthesis to cellular energy supplyProc Natl Acad Sci USA 1061778117786
  43. 43. HormanS.MorelN.VertommenD.et al.2008AMP-activated Protein Kinase Phosphorylates and Desensitizes Smooth Muscle Myosin Light Chain KinaseJ Biol Chem 2831850518512
  44. 44. HuG.JousilahtiP.BidelS.AntikainenR.TuomilehtoJ.2007Type 2 Diabetes and the Risk of Parkinson’s Disease.Diabetes Care30842847
  45. 45. InokiK.ZhuT.GuanK. L.2003TSC2 Mediates Cellular Energy Response to Control Cell Growth and Survival.Cell 115577590
  46. 46. InoueE.YamauchiJ.2006AMP-activated protein kinase regulates PEPCK gene expression by direct phosphorylation of a novel zinc finger transcription factorBBRC351793799
  47. 47. IxJ. H.SharmaK.2010Mechanisms Linking Obesity, Chronic Kidney Disease, and Fatty Liver Disease: The Roles of Fetuin-A, Adiponectin, and AMPKJ Am Soc Nephrol 21406412
  48. 48. JagerS.HandschinC.StPierreJ.SpiegelmanB. M.2007AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1alpha.Proc Natl Acad Sci USA 1041201712022
  49. 49. JakobsenS. N.HardieD. G.MorriceN.TornqvistH. E.2001AMP-activated Protein Kinase Phosphorylates IRS-1 on Ser-789 in Mouse C2C12 Myotubes in Response to 5-Aminoimidazole-4-carboxamide Riboside. J Biol Chem 2764691246916
  50. 50. JordanS. D.KonnerA. C.BruningJ. C.2010Sensing the fuels: glucose and lipid signaling in the CNS controlling energy homeostasisCMLS 6732553273
  51. 51. KawaguchiT.OsatomiK.YamashitaH.KabashimaT.UyedaK.2002Mechanism for Fatty Acid "Sparing" Effect on Glucose-induced Transcription. J Biol Chem 27738293835
  52. 52. KazganN.WilliamsT.ForsbergL. J.BrenmanJ. E.2010Identification of a Nuclear Export Signal in the Catalytic Subunit of AMP-activated Protein KinaseMol Biol Cell 2134333442
  53. 53. KimA. S.MillerE. J.YoungL. H.2009aAMP-activated protein kinase: a core signalling pathway in the heart. Acta Physiologica1963753
  54. 54. KimK.BaekA.HwangJ.E.ChoiY. A.JeongJ.LeeM.S.ChoD. H.LimJ.S.KimK. I.YangY.2009bAdiponectin-Activated AMPK Stimulates Dephos-phorylation of AKT through Protein Phosphatase 2A Activation. Cancer Res 6940184026
  55. 55. KimJ. H.ParkJ.M.YeaK.KimH. W.SuhP.G.RyuS. H.2010Phospholipase D1 Mediates AMP-Activated Protein Kinase Signaling for Glucose Uptake.PLoS ONEe9600 EOF
  56. 56. KingJ. D.Jr FitchA. C.LeeJ. K.Mc CaneJ. E.MakD. O.FoskettJ. K.HallowsK. R.2009AMP-activated protein kinase phosphorylation of the R domain inhibits PKA stimulation of CFTRAm J Physiol-Cell Physiol 297, C94101
  57. 57. Kodiha, M., Rassi, J. G., Brown, C. M. & Stochaj, U. (2007). Localization of AMP kinase is regulated by stress, cell density, and signaling through the MEK-->ERK1/2 pathway. Am J Physiol-Cell Physiol 293, C1427-1436.
  58. 58. KongsupholP.CassidyD.HiekeB.TreharneK. J.SchreiberR.MehtaA.KunzelmannK.2009Mechanistic insight into control of CFTR by AMPK. J Biol Chem 28456455653
  59. 59. KooS.H.FlechnerL.QiL.et al.2005The CREB coactivator TORC2 is a key regulator of fasting glucose metabolism. Nature 43711091111
  60. 60. KuramotoN.WilkinsM. E.FairfaxB. P.et al.2007Phospho-Dependent Functional Modulation of GABA(B) Receptors by the Metabolic Sensor AMP-Dependent Protein Kinase. Neuron 53233247
  61. 61. LageR.DiéguezC.Vidal-PuigA.LópezM.2008AMPK: a metabolic gauge regulating whole-body energy homeostasis. Trends Mol Med 14539549
  62. 62. LamiaK. A.SachdevaU. M.Di TacchioL.et al.2009AMPK Regulates the Circadian Clock by Cryptochrome Phosphorylation and Degradation. Science 326437440
  63. 63. LeclercI.SunG.MorrisC.Fernandez-MillanE.NyirendaM.RutterG.2011AMP-activated protein kinase regulates glucagon secretion from mouse pancreatic alpha cells. Diabetologia 54125134
  64. 64. LeeM.J.FeliersD.MariappanM. M.SataranatarajanK.et al.2007A role for AMP-activated protein kinase in diabetes-induced renal hypertrophy. Am J Physiol- Renal Physiol 292, F617F627.
  65. 65. LiJ.Mc CulloughL. D.2010Effects of AMP-activated protein kinase in cerebral ischemia. J Cereb Blood Flow Metab 30480492
  66. 66. LiY.RouxC. l.LazeregS.Le CaerJ.P.LapravoteO.BadetB.Badet-DenisotM.A.2007Identification of a Novel Serine Phosphorylation Site in Human Glutamine:Fructose-6-phosphate Amidotransferase Isoform 1 Biochem 461316313169
  67. 67. LiangJ.ShaoS. H.XuZ.X.HennessyB.DingZ.LarreaM.KondoS.DumontD. J.GuttermanJ. U.WalkerC. L.SlingerlandJ. M.MillsG. B.2007The energy sensing LKB1-AMPK pathway regulates 27kip1phosphorylation mediating the decision to enter autophagy or apoptosis. Nat Cell Biol 9, 218-224.
  68. 68. LieberthalW.LevineJ. S.2009The Role of the Mammalian Target Of Rapamycin (mTOR) in Renal Disease. J Am Soc Nephrol 2024932502
  69. 69. LippaiM.CsikosG.MaroyP.LukacsovichT.JuhaszG.SassM.2008SNF4Agamma, the Drosophila AMPK gamma subunit is required for regulation of developmental and stress-induced autophagy. Autophagy 4476486
  70. 70. LopaschukG. D.2008AMP-activated protein kinase control of energy metabolism in the ischemic heart. Int J Obesity 32, S29S35.
  71. 71. LuJ.WuD. M.ZhengY. L.HuB.ZhangZ. F.ShanQ.ZhengZ. H.LiuC. M.WangY. J.2010Quercetin activates AMP-activated protein kinase by reducing 2Cexpression protecting old mouse brain against high cholesterol-induced neurotoxicity. J Pathology 222, 199-212.
  72. 72. MaedaN.TakahashiM.FunahashiT.et al.2001PPAR Ligands Increase Expression and Plasma Concentrations of Adiponectin, an Adipose-Derived Protein. Diabetes 5020942099
  73. 73. MaherP. A.SchubertD. R.2009Metabolic links between diabetes and Alzheimer’s disease. Expert Rev Neurotherapeutics 9617630
  74. 74. MahlapuuM.JohanssonC.LindgrenK.et al.2004Expression profiling of the gamma-subunit isoforms of AMP-activated protein kinase suggests a major role for gamma3 in white skeletal muscle. Am J Physiol- Endocrinol Metabol 286, E194E200.
  75. 75. MairW.MorantteI.RodriguesA. P. C.ManningG.MontminyM.ShawR. J.DillinA.2011Lifespan extension induced by AMPK and calcineurin is mediated by CRTC-1 and CREB. Nature 470404408
  76. 76. MantovaniJ.RoyR.2011Re-evaluating the general(ized) roles of AMPK in cellular metabolism. FEBS Letters 585967972
  77. 77. MarsinA.S.BouzinC.BertrandL.HueL.2002The Stimulation of Glycolysis by Hypoxia in Activated Monocytes Is Mediated by AMP-activated Protein Kinase and Inducible 6-Phosphofructo-2-kinase. J Biol Chem 2773077830783
  78. 78. MarsinA. S.BertrandL.RiderM. H.et al.2000Phosphorylation and activation of heart PFK-2 by AMPK has a role in the stimulation of glycolysis during ischaemia. Curr Biol 1012471255
  79. 79. Mc DonaldA.FogartyS.LeclercI.HillE. V.HardieD. G.RutterG. A.2009Control of insulin granule dynamics by AMPK dependent KLC1 phosphorylation. Islets 1198209
  80. 80. Mc DonaldA.FogartyS.LeclercI.HillE. V.HardieD. G.RutterG. A.2010Cell-wide analysis of secretory granule dynamics in three dimensions in living pancreatic beta-cells: evidence against a role for AMPK-dependent phosphorylation of KLC1 at Ser517/Ser520 in glucose-stimulated insulin granule movement. Biochem Soc Transactions 38205208
  81. 81. Mc GeeS. L.HowlettK. F.StarkieR. L.Cameron-SmithD.KempB. E.HargreavesM.2003Exercise Increases Nuclear AMPK alpha2 in Human Skeletal Muscle. Diabetes 52926928
  82. 82. Mc GeeS. L.van DenderenB. J. W.HowlettK. F.MollicaJ.SchertzerJ. D.KempB. E.HargreavesM.2008AMP-Activated Protein Kinase Regulates GLUT4 Transcription by Phosphorylating Histone Deacetylase 5. Diabetes 57860867
  83. 83. Mc MahonK. W.ZanescuD. I.SoodV.PrabhakarS. S.2009The Role of 5’-AMP-Activated Protein Kinase (AMPK) in Diabetic Nephropathy: A New Direction? Curr Enzyme Inhibition 54450
  84. 84. MirouseV.SwickL. L.KazganN.StJohnston. D.BrenmanJ. E.2007LKB1 and AMPK maintain epithelial cell polarity under energetic stress. J Cell Biol 177387392
  85. 85. MiyamotoT.OshiroN.YoshinoK.NakashimaA.EguchiS.TakahashiM.OnoY.KikkawaU.YonezawaK.2008AMP-activated protein kinase phosphorylates Golgi-specific brefeldin A resistance factor 1 at Thr1337 to induce disassembly of Golgi apparatus. J Biol Chem 28344304438
  86. 86. NagataD.HirataY.2010The role of AMP-activated protein kinase in the cardio-vascular system. Hypertens Res 332228
  87. 87. NarbonneP.RoyR.2009Caenorhabditis elegans dauers need LKB1/AMPK to ration lipid reserves and ensure long-term survival. Nature 457210214
  88. 88. NieslerC. U.MyburghK. H.MooreF.2007The changing AMPK expression profile in differentiating mouse skeletal muscle myoblast cells helps confer increasing resistance to apoptosis. Experim Physiol 92207217
  89. 89. OakhillJ. S.ChenZ. P.ScottJ. W.SteelR.CastelliL. A.LingN.MacaulayS. L.KempB. E.2010beta-Subunit myristoylation is the gatekeeper for initiating metabolic stress sensing by AMP-activated protein kinase (AMPK). Proc Natl Acad Sci USA 1071923719241
  90. 90. PolekhinaG.GuptaA.MichellB. J.et al.2003AMPK β Subunit Targets Metabolic Stress Sensing to Glycogen. Curr Biol 13867871
  91. 91. PulinilkunnilT.HeH.KongD.AsakuraK.PeroniO. D.LeeA.KahnB. B.2011Adrenergic Regulation of AMP-activated Protein Kinase in Brown Adipose Tissue in Vivo. J Biol Chem 28687988809
  92. 92. PutmanC. T.MartinsK. J. B.GalloM. E.LopaschukG. D.PearceyJ. A.MacLean. I. M.SaranchukR. J.PetteD.2007alpha-Catalytic subunits of 5’AMP-activated protein kinase display fiber-specific expression and are upregulated by chronic low-frequency stimulation in rat muscle. Am J Physiology-Regul Integr Comp Physiol 293, R1325R1334.
  93. 93. QiJ.GongJ.ZhaoT.ZhaoJ.LamP.YeJ.LiJ. Z.WuJ.ZhouH.M.LiP.2008Downregulation of AMP-activated protein kinase by Cidea-mediated ubiquitination and degradation in brown adipose tissue. EMBO J 2715371548
  94. 94. QuanX.YuJ.BusseyH.StochajU.2007The localization of nuclear exporters of the importin-beta family is regulated by Snf1 kinase, nutrient supply and stress. Biochim Biophys Acta-Mol Cell Res 177310521061
  95. 95. QuentinT.KitzJ.SteinmetzM.PoppeA.BärK.KrätznerR.2011Different expression of the catalytic alpha subunits of the AMP activated protein kinase- an immunohistochemical study in human tissue. Histol Histopathol 26589596
  96. 96. RachidZ.KatsoulasA.WilliamsC.LarroqueA.L.Mc NameeJ.Jean-ClaudeB. J.2007Optimization of novel combi-molecules: Identification of balanced and mixed bcr-abl/DNA targeting properties. Bioorg Med Chem Lett 1742484253
  97. 97. ReihillJ. A.EwartM. A.HardieD. G.SaltI. P.2007AMP-activated protein kinase mediates VEGF-stimulated endothelial NO production. BBRC 35410841088
  98. 98. RonnettG. V.RamamurthyS.KlemanA. M.LandreeL. E.AjaS.2009AMPK in the brain: its roles in energy balance and neuroprotection. J Neurochem 1091723
  99. 99. RossF. A.RaffertyJ. N.DallasM. L.et al.2011Selective Expression in Carotid Body Type I Cells of a Single Splice Variant of the Large Conductance Calcium- and Voltage-activated Potassium Channel Confers Regulation by AMP-activated Protein Kinase. J Biol Chem 2861192911936
  100. 100. SaltI.CellerJ. W.HawleyS. A.PrescottA.WoodsA.CarlingD.HardieD. G.1998AMP-activated protein kinase: greater AMP dependence, and preferential nuclear localization, of complexes containing the alpha2 isoform. Biochem J 334177187
  101. 101. SandersM. J.GrondinP. O.HegartyB. D.SnowdenM. A.CarlingD.2007Investigating the mechanism for AMP activation of the AMP-activated protein kinase cascade. Biochem J 403139148
  102. 102. SchernhammerE.HansenJ.RugbjergK.WermuthL.RitzB.2011Diabetes and the Risk of Developing Parkinson’s Disease in Denmark. Diabetes Care, in press.
  103. 103. SchrauwenP.HesselinkM. K. C.2004Oxidative Capacity, Lipotoxicity, and Mitochondrial Damage in Type 2 Diabetes. Diabetes 5314121417
  104. 104. ShackelfordD. B.ShawR. J.2009The LKB1-AMPK pathway: metabolism and growth control in tumour suppression. Nat Rev Cancer 9563575
  105. 105. SkuratA. V.DietrichA. D.RoachP. J.2000Glycogen synthase sensitivity to insulin and glucose-6-phosphate is mediated by both NH2- and COOH-terminal phosphorylation sites. Diabetes 4910961100
  106. 106. SpasicM.CallaertsP.NorgaK. K.2009AMP-Activated Protein Kinase (AMPK) Molecular Crossroad for Metabolic Control and Survival of Neurons. Neuroscientist 15309316
  107. 107. SprenkleA. B.DaviesS. P.CarlingD.HardieD. G.SturgillT. W.1997Identification of Raf-1 Ser621 kinase activity from NIH3T3 cells as AMP-activated protein kinase. FEBS Letters 403254258
  108. 108. StapletonD.MitchelhillK. I.GaoG.et al.1996Mammalian AMP-activated Protein Kinase Subfamily. J Biol Chem 271611614
  109. 109. SteinS. C.WoodsA.JonesN. A.DavisonM. D.CarlingD.2000The regulation of AMP-activated protein kinase by phosphorylation. Biochem. J. 345437443
  110. 110. SteinbergG. R.KempB. E.2009AMPK in Health and Disease. Physiol Rev 8910251078
  111. 111. SteinbergG. R.MichellB. J.van DenderenB. J. W.et al.2006Tumor necrosis factor alpha-induced skeletal muscle insulin resistance involves suppression of AMP-kinase signaling. Cell Metabolism 4465474
  112. 112. SteinbergG. R.RushJ. W. E.DyckD. J.2003AMPK expression and phosphorylation are increased in rodent muscle after chronic leptin treatment. Am J Physiol-Endocrin Metabol 284, E648E654.
  113. 113. SteinbergG. R.WattM. J.FebbraioM. A.2009Cytokine regulation of AMPK signalling. Front Biosci 1419021916
  114. 114. SunW.LeeT.S.ZhuM.GuC.WangY.ZhuY.ShyyJ. Y.J.2006Statins Activate AMP-Activated Protein Kinase In Vitro and In Vivo. Circulation 11426552662
  115. 115. SuzukiA.OkamotoS.LeeS.SaitoK.ShiuchiT.MinokoshiY.2007Leptin Stimulates Fatty Acid Oxidation and Peroxisome Proliferator-Activated Receptor α Gene Expression in Mouse C2C12 Myoblasts by Changing the Subcellular Localization of the α2 Form of AMP-Activated Protein Kinase. Mol Cell Biol 2743174327
  116. 116. TakiarV.NishioS.Seo-MayerP.KingJ. D.LiH.ZhangL.KarihalooA.HallowsK. R.SomloS.CaplanM. J.2011Activating AMP-activated protein kinase (AMPK) slows renal cystogenesis. Proc Natl Acad Sci USA 10824622467
  117. 117. ThorntonC.BrightN. J.SastreM.MuckettP. J.CarlingD.2011AMP-activated protein kinase (AMPK) is a tau kinase, activated in response to amyloid beta-peptide exposure. Biochem J 434503512
  118. 118. TowlerM. C.HardieD. G.2007AMP-activated protein kinase in metabolic control and insulin signaling. Circulation Res 10032841
  119. 119. TreebakJ. T.TaylorE. B.WitczakC. A.AnD.ToyodaT.KohH.J.XieJ.FeenerE. P.WojtaszewskiJ. r. F. P.HirshmanM. F.GoodyearL. J.2010Identification of a novel phosphorylation site on TBC1D4 regulated by AMP-activated protein kinase in skeletal muscle. Am J Physiol-Cell Physiol 298, C377385
  120. 120. TurnleyA. M.StapletonD.MannR. J.WittersL. A.KempB. E.BartlettP. F.1999Cellular Distribution and Developmental Expression of AMP-Activated Protein Kinase Isoforms in Mouse Central Nervous System. J Neurochem 7217071716
  121. 121. TzatsosA.TsichlisP. N.2007Energy Depletion Inhibits Phosphatidylinositol 3-Kinase/Akt Signaling and Induces Apoptosis via AMP-activated Protein Kinase-dependent Phosphorylation of IRS-1 at Ser-794. J Biol Chem 2821806918082
  122. 122. UmJ. H.YangS.YamazakiS.KangH.ViolletB.ForetzM.ChungJ. H.2007Activation of 5’-AMP-activated Kinase with Diabetes Drug Metformin Induces Casein Kinase 1ε (CKIε)-dependent Degradation of Clock Protein mPer2. J Biol Chem 2822079420798
  123. 123. van OortM. M.van DoornJ. M.HasnaouiM. E.GlatzJ. F.BonenA.van der HorstD. J.RodenburgK. W.J. J. P. L.2009Effects of AMPK activators on the sub-cellular distribution of fatty acid transporters CD36 and FABPpm. Arch Physiol Biochem 115137146
  124. 124. VerniaS.Solaz-FusterM. C.Gimeno-AlcanizJ. V.et al.2009AMP-activated Protein Kinase Phosphorylates R5/PTG, the Glycogen Targeting Subunit of the R5/PTG-Protein Phosphatase 1 Holoenzyme, and Accelerates Its Down-regulation by the Laforin-Malin Complex. J Biol Chem 28482478255
  125. 125. VigettiD.ClericiM.DeleonibusS.KarousouE.ViolaM.MorettoP.HeldinP.HascallV. C.De LucaG.PassiA.2011Hyaluronan Synthesis Is Inhibited by Adenosine Monophosphate-activated Protein Kinase through the Regulation of HAS2 Activity in Human Aortic Smooth Muscle Cells. J Biol Chem 28679177924
  126. 126. VingtdeuxV.DaviesP.DicksonD.MarambaudP.2011AMPK is abnormally activated in tangle- and pre-tangle-bearing neurons in Alzheimer’s disease and other tauopathies. Acta Neuropathol 121337349
  127. 127. ViolletB.AtheaY.MounierR.GuigasB.ZarrinpashnehE.HormanS.LantierL.HebrardS.Devin-LeclercJ.BeauloyeC.ForetzM.AndreelliF.Ventura-ClapierR.BertrandL.2009aAMPK: Lessons from transgenic and knockout animals. Front Biosc 141944
  128. 128. ViolletB.LantierL.Devin-LeclercJ.HebrardS.AmouyalC.MounierR.ForetzM.AndreelliF.2009bTargeting the AMPK pathway for the treatment of Type 2 diabetes. Front Biosci 1433803400
  129. 129. ViolletB.HormanS.LeclercJ.LantierL.ForetzM.BillaudM.GiriS.AndreelliF.2010AMPK inhibition in health and disease. Crit Rev Biochem Mol Biol 45276295
  130. 130. WangA.LiuM.LiuX.DongL. Q.GlickmanR. D.SlagaT. J.ZhouZ.LiuF.2011Up-regulation of adiponectin by resveratrol: The essential roles of the Akt/FOXO1 and AMPK signaling pathways and DsbA-L. J Biol Chem 2866066
  131. 131. WangT.YuQ.ChenJ.DengB.QianL.Le Y.20102AMediated AMPK Inhibition Promotes HSP70 Expression in Heat Shock Response. PLoS ONE 5, e13096.
  132. 132. WangW.YangX.KawaiT.de SilanesI. L. p.Mazan-MamczarzK.ChenP.ChookY. M.QuenselC.ÃhlerK.M.GorospeM.2004AMP-activated Protein Kinase-regulated Phosphorylation and Acetylation of Importin α1. J Biol Chem 2794837648388
  133. 133. WardenS. M.RichardsonC.O’DonnellJ.StapletonD.KempB. E.WittersL. A.2001Post-translational modifications of the beta-1 subunit of AMP-activated protein kinase affect enzyme activity and cellular localization. Biochem J 354275283
  134. 134. WattM. J.HolmesA. G.PinnamaneniS. K.GarnhamA. P.SteinbergG. R.KempB. E.FebbraioM. A.2006Regulation of HSL serine phosphorylation in skeletal muscle and adipose tissue. Am J Physiol-Endocrin Metabol 290, E500E508.
  135. 135. WitczakC.SharoffC.GoodyearL.2008AMP-activated protein kinase in skeletal muscle: From structure and localization to its role as a master regulator of cellular metabolism. CMLS 6537373755
  136. 136. XieM.ZhangD.DyckJ. R. B.et al.2006A pivotal role for endogenous TGF-beta-activated kinase-1 in the LKB1/AMP-activated protein kinase energy-sensor pathway. Proc Natl Acad Sci USA 1031737817383
  137. 137. YangW.HongY. H.ShenX.Q.FrankowskiC.CampH. S.LeffT.2001Regulation of Transcription by AMP-activated Protein Kinase. J Biol Chem 2763834138344
  138. 138. ZhangB. B.ZhouG.LiC.2009AMPK: An Emerging Drug Target for Diabetes and the Metabolic Syndrome. Cell Metabolism 9407416
  139. 139. ZhengD.PerianayagamA.LeeD. H.BrannanM. D.YangL. E.TellalianD.ChenP.LemieuxK.MaretteA.YounJ. H.Mc DonoughA. A.2008AMPK activation with AICAR provokes an acute fall in plasma [K+]. Am J Physiol-Cell Physiol 294, C126C135.

Written By

Mohamed Kodiha and Ursula Stochaj

Submitted: 10 November 2010 Published: 12 September 2011