Skip to main content
Acta Pharmaceutica Sinica. B logoLink to Acta Pharmaceutica Sinica. B
. 2015 Jul 21;6(1):1–19. doi: 10.1016/j.apsb.2015.06.002

Regulation of AMP-activated protein kinase by natural and synthetic activators

David Grahame Hardie 1,
PMCID: PMC4724661  PMID: 26904394

Abstract

The AMP-activated protein kinase (AMPK) is a sensor of cellular energy status that is almost universally expressed in eukaryotic cells. While it appears to have evolved in single-celled eukaryotes to regulate energy balance in a cell-autonomous manner, during the evolution of multicellular animals its role has become adapted so that it also regulates energy balance at the whole body level, by responding to hormones that act primarily on the hypothalamus. AMPK monitors energy balance at the cellular level by sensing the ratios of AMP/ATP and ADP/ATP, and recent structural analyses of the AMPK heterotrimer that have provided insight into the complex mechanisms for these effects will be discussed. Given the central importance of energy balance in diseases that are major causes of morbidity or death in humans, such as type 2 diabetes, cancer and inflammatory disorders, there has been a major drive to develop pharmacological activators of AMPK. Many such activators have been described, and the various mechanisms by which these activate AMPK will be discussed. A particularly large class of AMPK activators are natural products of plants derived from traditional herbal medicines. While the mechanism by which most of these activate AMPK has not yet been addressed, I will argue that many of them may be defensive compounds produced by plants to deter infection by pathogens or grazing by insects or herbivores, and that many of them will turn out to be inhibitors of mitochondrial function.

KEY WORDS: AMP-activated protein, kinase, Energy balance, AMP, AMPK activator, Mitochondrial function, Regulatory mechanism

Graphical abstract

AMPK monitors energy balance at the cellular level by sensing the ratios of AMP/ATP and ADP/ATP, and recent structural analyses of the AMPK heterotrimer that have provided insight into the complex mechanisms for these effects will be discussed.

graphic file with name fx1.jpg

1. Introduction

The 5'-adenosine monophosphate (AMP)-activated protein kinase (AMPK) is a sensor of cellular energy that helps to maintain energy balance at both the cellular and whole body levels1, 2, 3, 4. Since type 2 diabetes, which affects 5%−10% of the world population, can be regarded as a disorder of energy balance caused by over-nutrition, there has been much interest in AMPK as a drug target. It is also becoming apparent that two other major causes of human death and morbidity, i.e., cancer and inflammatory disease, can be viewed as metabolic derangements. Thus, tumor cells and cells, involved in inflammation, tend to display a glycolytic phenotype (termed the Warburg effect or aerobic glycolysis), whereas quiescent cells and cells involved in the resolution of inflammatory responses tend to utilize oxidative metabolism5. Since AMPK inhibits cell growth and proliferation, and also promotes the more glucose-sparing and energy-efficient mitochondrial oxidative metabolism rather than glycolysis, interest in the system as a drug target in the fields of cancer and inflammatory disease has been steadily increasing.

Following its initial definition by our group in the late 1980s6, 7, 8, over 9000 papers have been published on the AMPK system, and it is not possible to give a full coverage of the field in a single article. In this review I will focus on its structure and evolution, its regulation by metabolites, and its modulation by synthetic compounds that are being developed as pharmacological AMPK activators and by natural products that are being tested as medicines.

2. AMPK—subunit structure and evolution

AMPK appears to exist in almost all eukaryotic species as heterotrimeric complexes comprising a catalytic α subunit and regulatory β and γ subunits. In humans and other mammals, the α subunits are encoded by two genes (PRKAA1/PRKAA2, encoding α1/α2), the β subunits by two (PRKAB1/PRKAB2, encoding β1/β2) and the γ subunits by three (PRKAG1/PRKAG2/PRKAG3, encoding γ1/γ2/γ3). All twelve combinations of α, β and γ subunit isoforms are able to form heterotrimeric complexes when co-expressed, although certain combinations appear to be favored in vivo9. Genes encoding orthologs of AMPK-α, -β and -γ subunits are readily found in all eukaryotes where genome sequences have been completed. The one known exception to this is the microsporidian Encephalitozoon cuniculi, an obligate intracellular parasite that lives inside other mammalian cells including those of humans, and which has no free-living form other than metabolically inert spores10. While a genuine eukaryote, E. cuniculi has an extremely small genome encoding only 29 conventional protein kinase catalytic subunits, and lacks genes encoding the α, β and γ subunits of AMPK11. It does contain genes encoding the enzymes required for a complete glycolytic pathway10, but lacks adenosine-triphosphate (ATP)-generating mitochondria although having mitochondrial remnants termed mitosomes12. Interestingly, E. cuniculi expresses unusual transmembrane ATP/adenosine diphosphate (ADP) translocases, some of which appear to be located in the plasma membrane13. The implication of this is that the organism may utilize these translocases to “steal” ATP from the host cell in exchange for ADP. E. cuniculi may therefore have been able to afford to lose genes encoding AMPK, because its host cell does express the kinase and can regulate energy homeostasis on its behalf.

Given that AMPK is found in essentially all present day eukaryotes, it seems likely that it evolved soon after the development of the first eukaryote. It is widely believed that the key event that led to the first eukaryotic cell was the endosymbiotic acquisition by an archaeal host cell of aerobic bacteria, which eventually became mitochondria. One can speculate that the host cell would have needed a system to monitor the output of their newly acquired oxidative organelles, and to regulate the ability of those organelles to supply ATP according to the demands of the host. AMPK fits the bill to be such a system: for example, in the budding yeast Saccharomyces cerevisiae the AMPK ortholog is not required for growth by the fermentative metabolism (i.e., glycolysis) that is utilized in high glucose, but is required for the switch to oxidative metabolism that occurs when glucose run low14. Similarly, mitochondrial biogenesis is one of the key downstream effects of AMPK activation in mammalian cells15, 16, 17.

Most energy-requiring processes in eukaryotic cells are driven, either directly or indirectly, by hydrolysis of ATP to ADP, and it is possible to draw an analogy between these nucleotides and the chemicals in a rechargeable battery. A high ratio of ATP to ADP is equivalent to a fully charged battery, while if this ratio is falling the cellular battery is becoming flat. Extending this analogy, AMPK can be regarded as the biological equivalent of the system within a cell-phone or laptop computer that monitors the battery charge. As discussed in more detail in Section 3, it is activated by increasing ratios of AMP/ATP and ADP/ATP. An increase in either ratio signifies falling cellular energy, but if the reversible reaction catalyzed by adenylate kinase (2ADP↔ATP+AMP) is at equilibrium (as seems to be the case in most eukaryotic cells) it is easy to show that the AMP/ATP ratio will vary as the square of the ADP/ATP ratio18, making the former a much more sensitive signal of falling energy status than the latter. A full description of the downstream targets for AMPK is beyond the scope of this article, and readers interested in that aspect should consult other reviews (e.g., Ref. 19). However, once activated by energy stress, AMPK attempts to restore cellular energy homeostasis by activating catabolic pathways that generate ATP, while switching off ATP-consuming processes not essential to short-term cell survival, including almost all anabolic pathways. Although AMPK almost certainly arose in single-celled eukaryotes as a cell-autonomous mediator of energy balance, it is intriguing that role of the system seems to have become adapted during the evolution of multicellular eukaryotes so that it also regulates energy balance at the whole body level. It does this particularly by mediating effects of hormones acting on the hypothalamus of the brain that control energy intake (i.e., feeding) and energy expenditure1, 2, 3, 4.

3. Canonical regulation by phosphorylation and by adenine nucleotides

AMPK is normally only significantly active after phosphorylation of a conserved threonine residue within the activation loop of the kinase domain on the α subunit. This threonine residue is usually referred to as Thr172 due to its position in the rat α2 subunit where originally identified20, although the precise numbering may differ in other isoforms and species. Following a long search, the primary upstream kinase that phosphorylates Thr172 in vivo was shown to be a heterotrimeric complex between the tumor suppressor kinase liver kinase B1 (LKB1), the pseudokinase STE20-related adaptor (STRAD) and the scaffold protein mouse protein 25 (MO25)21, 22, 23. This complex appears to be constitutively active in that its activity is not regulated under situations of energy stress when AMPK is activated in an LKB1-dependent manner24, 25. Nevertheless, binding of AMP to AMPK can regulate both the phosphorylation of Thr172 by LKB1, and its dephosphorylation (see below). Almost as soon as it was found that LKB1 was the primary upstream kinase, it was realized that there was some phosphorylation of Thr172 even in tumor cells that had lost LKB1, and this was traced to the calmodulin-dependent protein kinase, calcium/calmodulin-dependent protein kinase kinase β (CaMKKβ)26, 27, 28. This provides an alternate Ca2+-mediated upstream pathway for AMPK activation, which mediates effects of hormones and mediators acting through Gq/G11-coupled receptors that trigger release of Ca2+ from intracellular stores via the second messenger inositol-1,4,5-trisphosphate (IP3)29. Such hormones include thrombin acting on endothelial cells via the protease-activated receptor30, and ghrelin acting on hypothalamic neurons via the glutathione reductase 1 (GSHR1) receptor31. Thr172 can also be phosphorylated, and AMPK activated, in intact cells by the protein kinase transforming growth factor-β-activated kinase-1 (TAK1)32, 33, although the physiological relevance of that mechanism currently remains unclear.

Allosteric activation of the phosphorylated kinase by 5′-AMP was originally demonstrated in 198034 (before AMPK acquired its current name), but in the early 1990s it was shown that AMP binding to AMPK not only caused allosteric activation but also promoted its net phosphorylation at Thr17235. It is now clear that AMP binding has three effects on AMPK36 that activate the system in a synergistic manner, making the final response very sensitive to even small changes in AMP:

  • (i)

    promotion of phosphorylation by LKB1, but not CaMKKβ (although this selectivity for LKB1 has been disputed37);

  • (ii)

    protection against dephosphorylation of Thr172 by protein phosphatases; and

  • (iii)

    allosteric activation of the phosphorylated kinase.

Of these three effects, it has been reported that mechanisms (i)37 and (ii)38 are also mimicked by binding of ADP. Given that ADP is present in unstressed cells at concentrations ten times higher than AMP, and that allosteric activation (which is only caused by AMP binding) is often reported as being small in magnitude (<2-fold), this led to proposals that ADP rather than AMP might be the crucial activator of AMPK37, 38, 39. However, our group36 reported that while mechanism (ii) can indeed be caused by binding of ADP, AMP is about 10-fold more potent. Moreover, using a native preparation of mammalian AMPK rather than a bacterially expressed complex, allosteric activation by AMP can be substantial (>10-fold), even in the presence of concentrations of ATP that are 1−2 orders of magnitude higher and within the physiological range (5 mmol/L)36. Thus, while ADP may contribute to activation, we would argue that AMP remains the primary regulator of AMPK.

4. Pharmacological activators of AMPK

Since the realization in the late 1990s that activation of AMPK might be useful for treatment of type 2 diabetes40, numerous pharmacological activators have been developed. Based on their mechanism of action, they can be divided into four classes that are discussed in 4.1, 4.2, 4.3, 4.4.

4.1. Activators that act indirectly by inhibiting cellular ATP synthesis

Since depletion of ATP always causes increases in AMP and ADP, AMPK is activated by any compound that inhibits ATP synthesis. In cells that are primarily using glycolysis to generate ATP (as in most rapidly proliferating cells), AMPK is activated by inhibitors of glycolysis such as 2-deoxyglucose41. A much larger class of activators, some of which are shown in Fig. 1A, are those that inhibit mitochondrial ATP synthesis by inhibiting the respiratory chain at Complex I (e.g., metformin or phenformin42, 43) or Complex III (e.g., antimycin A44), or that inhibit Complex V, the mitochondrial F1 ATP synthase (e.g., oligomycin or resveratrol41, 45). All of these agents will increase cellular ADP/ATP and/or AMP/ATP ratios, although correlations between such ratios and changes in AMPK activity do not prove on their own that activation by AMP or ADP is the sole mechanism. The best method to confirm this is to use a cell line expressing an AMP/ADP-insensitive mutant of AMPK such as the R531G mutation in γ241 or the equivalent R299G mutation in γ146. Any agent that activates AMPK solely by increasing the cellular levels of AMP or ADP will fail to activate such mutants. Further discrimination can be obtained by measuring cellular oxygen uptake and acidification of the medium using an extracellular flux analyzer. Compounds that inhibit mitochondrial function should inhibit oxygen uptake, while those that inhibit glycolysis should reduce lactate output and hence extracellular acidification. For example, the compound PT-1, which was originally proposed to act by direct binding to AMPK47, was recently shown using these methods to act instead by inhibiting the respiratory chain46.

Figure 1.

Figure 1

Structures of AMPK-activating compounds that act via: (A) inhibiting mitochondrial ATP synthesis; (B) pro-drugs converted to active agents inside cells, as shown; and (C) direct activators. (D) shows the mechanism by which antifolate drugs activate AMPK by causing accumulation of ZMP, an intermediate in the synthesis of the purine nucleotides inosine monophosphate (IMP), AMP and guanosine monophosphate (GMP).

In the last few years well over 100 natural products or extracts derived from plants, many of which are used in traditional Asian medicines, have been reported to activate AMPK. These are considered in more detail in Section 7. However, it is worth stating here that several of them, including berberine41, 48 and arctigenin49, appear to activate AMPK by inhibiting the mitochondrial respiratory chain, as does galegine, a natural product from the medicinal plant Galega officinalis from which metformin and phenformin were derived41, 50 (Fig. 1A). At least one potent synthetic compound, derived from a high-throughput screen designed to detect compounds that activate AMPK in cell-based assays, has also been shown to activate AMPK by inhibiting Complex I of the respiratory chain51.

4.2. Pro-drugs that are converted into AMP analogs inside cells

It is clear that the regulatory adenine nucleotide-binding sites on the γ subunits of AMPK, which are discussed in more detail below, require the presence of negatively charged phosphate groups on bound nucleotides, and it therefore may be difficult to develop cell-permeable AMP analogs that bind these sites. However, a related approach is to develop pro-drugs that are cell permeable but are converted following their uptake into AMP analogs by cellular enzymes. In fact, 5-aminoimidazole-4-carboxamide ribonucleoside, the first pharmacological AMPK activator to be developed49, 50, works by this mechanism. This compound is often referred to as AICAR and I adopt this usage below, although this can cause confusion because researchers in the field of nucleotide metabolism use the same acronym to describe the phosphorylated ribotide form, which I will refer to instead as ZMP (AICAR monophosphate). AICAR is an adenosine analog that is taken up into cells by adenosine transporters52 and phosphorylated by intracellular adenosine kinase into ZMP (Fig. 1B). ZMP is an AMP analog that binds to AMPK at the same sites as AMP53 and mimics all of the effects of AMP on the AMPK system54. In fact, ZMP has low potency compared with AMP54, but AICAR nevertheless activates AMPK in most primary cells and tissues because AICAR is rapidly converted to ZMP, which is then metabolized much more slowly. ZMP therefore accumulates within many cells to concentrations within the millimolar range (even higher than the external AICAR concentration), which is necessary for it to activate AMPK. It is important to note that ZMP is a natural intermediate in purine nucleotide synthesis, and some immortalized cell lines have a high rate of purine synthesis such that ZMP does not accumulate in response to extracellular AICAR, and AMPK is therefore not activated. Interestingly, however, antifolate drugs that are used to treat cancer, or inflammatory disorders such as rheumatoid arthritis, inhibit the transformylase that catalyzes the first step in the metabolism of ZMP to purine nucleotides, thus causing accumulation of ZMP (Fig. 1D). For example, the antifolate methotrexate dramatically sensitizes cells to the activating effects of AICAR55, while pemetrexed can activate AMPK even in the absence of exogenous AICAR56.

Recently, a synthetic compound that activates AMPK by a pro-drug mechanism has been developed. C13 is a phosphonate diester that is taken up into cells and converted by cellular esterases into C2 (Fig. 1B), an AMP analog that is 2−3 orders of magnitude more potent as an allosteric activator of AMPK than AMP, and 4 orders of magnitude more potent than ZMP57. Another major advantage of C13 over AICAR is that C2, unlike ZMP, does not modulate other AMP-sensitive enzymes such as glycogen phosphorylase, phosphofructokinase or fructose-1,6-bisphosphatase58. C2 is, however, selective for AMPK complexes containing the α1 rather than the α2 isoform58, an interesting finding that is considered in more detail in Section 5.3 below. Finally, 3'-deoxyadenosine (cordycepin) is a bioactive compound derived from the fungus Cordyceps militaris, which is an analog of adenosine lacking oxygen on the 3' position of the ribose ring. Although it has been shown to activate AMPK in intact cells and to bind directly to the AMPK-γ subunit59, 60, it is perhaps more likely that the true activator is cordycepin-5'-monophosphate generated from cordycepin within the cell.

4.3. Allosteric activators that bind directly to AMPK at sites distinct from the AMP sites

The first compound in this class was A-769662 (Fig. 1C), developed by Abbott laboratories from a high throughput screen searching for allosteric activators of purified AMPK. Although it has poor oral availability, when administered by intraperitoneal injection it was found to have favorable effects on the metabolism of an insulin-resistant animal model, the ob/ob mouse61. A-769662 did not increase cellular ADP/ATP or AMP/ATP ratios61, still activated AMPK in cells expressing an AMP-insensitive mutant41, and did not displace AMP from its binding sites on the γ subunit62, suggesting that it bound at a different site from AMP even though, like AMP, it caused both allosteric activation and protection against Thr172 dephosphorylation62, 63. A-769662 is also selective for activation of β1 rather than β2 complexes64, and its effects are abolished by an S108A mutation in β1 that prevents the autophosphorylation of that serine residue63, suggesting that the binding site involved the β subunit. As discussed in Section 5.2 below, the binding site has now been identified by structural biology to be a cleft located between the N-lobe of the kinase domain on the α subunit and the carbohydrate-binding module on the β-subunit. Another, more potent activator that binds at this site, 99165 (also known as ex22966), has emerged from high-throughput screens (Fig. 1C). Like A-769662, this compound shows some selectivity for β1 complexes although it will activate β2 complexes at higher concentrations. A third compound, MT 63-7867 (Fig. 1C), also shows selectivity for β1 complexes and may therefore bind this site, although this has not yet been formally demonstrated. None of these compounds has yet entered clinical trials. However, it should be noted that, of these compounds, only A-769662 has been available for a prolonged period, and enthusiasm for its entry into clinical trials may have been dampened in part by poor oral availability61 and in part by the occurrence of AMPK-independent, “off-target” effects68.

A key question regarding the binding site for A-769662 is whether it binds any naturally occurring ligands. One natural product derived from plants that does bind to this site is salicylate69, which has been used as a medicinal compound by humans since ancient times70. Acetyl salicylic acid (ASA or aspirin), which is broken down to salicylate within minutes of its adsorption into the bloodstream, is a synthetic derivative developed in the 1890s as a less irritating formulation to deliver salicylate orally. Aspirin is a potent inhibitor of the cyclo-oxygenases71 (COX1 and COX2) that catalyze the key initial steps in the biosynthesis of prostaglandins and other eicosanoids; irreversible inhibition of synthesis of the eicosanoid thromboxane A2 in platelets is the mechanism by which it inhibits platelet aggregation and hence blood clotting72. However, since aspirin and salicylate have equal potency as anti-inflammatory agents, yet salicylate is a very poor COX inhibitor, it remains unclear whether all of the anti-inflammatory actions of aspirin can be attributed to COX inhibition73. In 2012 we reported that salicylate, but not aspirin, activated AMPK69. Like A-769662, salicylate is a poor activator of β2 complexes and its effect were abolished by an S108A mutation in β1, so it seemed likely that it bound to the same site as A-76966269, a proposal recently confirmed by a crystal structure of the human α1β1γ1 complex with bound iodosalicylate74. When salicylate or A-769662 were injected into wild type mice, they promoted a more rapid switch from carbohydrate to fat oxidation on food withdrawal, as would be expected for an AMPK activator that triggered phosphorylation and inactivation of both isoforms of acetyl-CoA carboxylase (ACC1 and ACC2) and hence caused a rapid switch from fat synthesis to fat oxidation. However, these effects were lost in AMPK-β1 knockout mice; since salicylate and A-769662 do not activate β2-containing complexes, this provided strong evidence that these metabolic effects were mediated by AMPK69.

When AMP and A-769662 are added to AMPK together, they cause a synergistic allosteric activation even of “naïve” AMPK complexes that are not phosphorylated on Thr172, although prior autophosphorylation of Ser108 (or a phosphomimetic S108E mutation) is required for a maximal effect, as well as for a maximal response to A-769662 alone75. Synergism between these activating sites may also be relevant in intact cells, because metformin (which increases cellular AMP) and salicylate act synergistically to activate AMPK and inhibit fat synthesis in isolated mouse and human hepatocytes—while little AMPK activation was observed with metformin or salicylate on their own at concentrations (100 µmol/L and 300−500 µmol/L, respectively) observed in human plasma following normal doses, significant effects were observed when they were given together69. There were also additive effects of low doses of metformin and salicylate in vivo to activate AMPK in livers of high-fat fed mice, accompanied by reduced liver triglycerides and increased hepatic insulin sensitivity69.

4.4. Oxidative stress

It was reported in 2001 that oxidative stress produced by hydrogen peroxide increased Thr172 phosphorylation and activated AMPK; this was accompanied by increases in AMP/ATP ratios, suggesting that the effect might be AMP-dependent (i.e., the mechanism described in Section 4.1)76. More recently, Zmijewski et al.77 used glucose oxidase to generate H2O2 from glucose present in the medium—this appears to be a better model for physiological oxidative stress, because it generates a constant low level of H2O2 in the medium (<20 µmol/L) rather than a transient spike of much higher concentrations that is obtained by adding H2O2 directly78. Zmijewski et al. reported that glucose oxidase treatment of HEK-293 cells did not cause decreases in ATP levels, and presented evidence that AMPK activation was caused instead by oxidation of two conserved cysteine residues (Cys299 and Cys304) present in the auto-inhibitory domain of the α subunit (see Section 5.1). However, our group78 reported that glucose oxidase treatment did increase AMP/ATP ratios in the same cell line, and that AMPK activation was largely abolished in HEK-293 cells expressing the AMP-insensitive R531G mutant of γ2. While this suggested that the effect was primarily AMP-dependent, there was a small residual effect observed with the R531G mutant that might be explained by the mechanism described by Zmijewski et al.77. More recently, Shao et al.79 reported that AMPK was inactivated rather than activated by oxidative stress in primary cardiomyocytes, and that this was prevented by thioredoxin. Inactivation was traced to oxidation of two cysteine residues within the kinase domain of AMPK (Cys130 and Cys174), distinct from those whose oxidation was proposed by Zmijewski et al.77 to cause activation of AMPK. Cys174 is almost adjacent to Thr172, and unmodified cysteine residues at these positions were shown to be necessary for activation by LKB1. Shao et al.79 suggested that the activation of AMPK caused by oxidative stress in HEK-293 cells77, 78 may occur because higher levels of anti-oxidant enzymes in this immortalized cell line may prevent the inactivation that they observed in primary cardiomyocytes.

4.5. Why do different pharmacological activators of AMPK have different effects?

Some of the pharmacological activators of AMPK discussed above have been used as medicines by humans for decades (metformin), centuries (berberine) or even millennia (salicylate). Why are their pharmacological effects so different? One potential explanation is pharmacokinetics—for example, metformin is a cation with poor cell permeability, and it requires expression of transporters of the organic cation transporter (OCT) family, such as OCT1, for cellular uptake. Because OCT1 is highly expressed in hepatocytes, 24% of an intravenous dose of metformin was found in the liver of wild type mice ten minutes after injection, compared with <1% in Oct1−/− knockout mice80. Thus, the effects of metformin in vivo are likely to be restricted to the liver, whereas other compounds will also activate AMPK in other organs or cell types. In addition, since metformin activates AMPK indirectly by inhibiting the respiratory chain and thus increasing cellular AMP and ADP41, it is likely that it has many “off-target” or AMPK-independent effects; indeed the acute effects of metformin on hepatic glucose production81, as opposed to its longer-term effects on hepatic insulin sensitivity82, appear to be independent of AMPK. Similarly, although salicylate does bind directly to AMPK, being a particularly small molecule it is unlikely to bind to any target with high affinity, and it almost certainly has several AMPK-independent effects. Acetyl salicylate (aspirin) is, of course, already known to inhibit cyclo-oxygenases and hence prostanoid biosynthesis, although salicylate itself is a relatively poor cyclo-oxygenase inhibitor83. The different pharmacological effects of these AMPK activators may therefore be due to a combination of different pharmacokinetics, and distinct AMPK-independent effects.

5. Domain architecture and structure of AMPK

5.1. The α subunits

Each AMPK-α subunit contains at the N-terminus a typical eukaryotic kinase domain, with a conventional small N-lobe consisting mainly of β-sheets, followed by the larger C-lobe consisting mainly of α-helices. In the most recent crystal structures38, 65, 74, 84, such as that shown in Fig. 2, the kinase had been crystallized in the presence of the non-specific, ATP-competitive kinase inhibitor staurosporine, and as expected this was located in the ATP-binding cleft between the N- and C-lobes. The critical phosphorylation site, Thr172, is located in the “activation loop”, a sequence region that must be phosphorylated in many kinases before they become active. Most of the crystal structures of AMPK were obtained with Thr172 phosphorylated and the activation loop was well ordered, although in at least one structure in the unphosphorylated state the activation loop was partially disordered84. The α subunit kinase domain (α-KD) is immediately followed by the auto-inhibitory domain (α-AID), so-called because bacterially expressed α-KD:α-AID constructs are about 10-fold less active than constructs containing the α-KD only, even when both have been phosphorylated on Thr17262, 85. There is now good evidence that the α-AID inhibits the α-KD when AMP is not bound to the γ subunit, thus explaining the 10-fold allosteric activation by AMP. Crystal structures of α-KD:α-AID constructs from the AMPK ortholog from the fission yeast Schizosaccharomyces pombe86, and more recently from humans84, show that in this low activity state the α-AID, a bundle of three short α-helices, binds to the α-KD on the opposite surface to the catalytic cleft, with the α3 helix of the α-AID interacting with the N-lobe and the hinge between the N- and C-lobes (Fig. 3A). By comparing many structures of kinase domains in active and inactive conformations, it has been found that four hydrophobic residues termed the “regulatory spine” are universally aligned in active conformations, indicating that the active site is correctly disposed for activity, but that these residues are out of alignment in inactive conformations87. In the structures of the inactive α-KD:α-AID constructs of AMPK, the four residues that form the “regulatory spine” (Leu68 and Leu79 from the N-lobe, and His137 and Phe158 from the C-lobe) are not aligned (Fig. 3A). By contrast, in all structures of AMPK heterotrimers in active states, which are phosphorylated on Thr172 and have AMP bound to the γ subunit (see below), the α-AID has undergone a rotation such that helix α3 now interacts primarily with the γ subunit rather than with the N-lobe of the α-KD. At the same time the α-KD switches to an active conformation, where the four residues of the regulatory spine are now stacked in alignment (Fig. 3B).

Figure 2.

Figure 2

Structure of complete α1β2γ1 heterotrimer of AMPK. The model was created with MacPyMol using PDB file 4RER84. All molecules are shown in “sphere view”, omitting hydrogen atoms. Domains of the heterotrimer are color coded and labeled as decribed in the text, whereas ancillary ligands (β-cyclodextrin, staurosporine and AMP) are shown with carbon atoms in light gray, oxygen in red and nitrogen in blue. AMP in site 3 is just visible beneath α-RIM2, while AMP in sites 1 and 4 are located around the other side of the γ1 subunit.

Figure 3.

Figure 3

Two views of the kinase and auto-inhibitory domains of the α subunit (α-KD:α-AID) in inactive (A) and active (B) conformations. Note the major rotation of the α-AID relative to the α-KD between the two models; in (A), α-AID helix α3 interacts mainly with the α-KD small lobe (and with the hinge between the small and large lobes), but in (B) it interacts mainly with the γ subunit (not shown) instead. Note also that the four side chains of the “regulatory spine” (in white, red, magenta and blue) are out of alignment in (A) but are stacked in alignment in (B), indicating an active conformation87. The models were created with MacPyMol using PDB files 4RED (A) and 4RER (B)84, and are shown in “cartoon” view except for the four residues that form the “regulatory spine” which are in “sphere” view. The view in (A) is of a structure derived from a construct containing only the α-KD and α-AID of human AMPK-α1. The structure crystallized as a dimer, and the α-KD shown is from one molecule while the α-AID shown is from the other molecule within the dimer. Nevertheless, the α-KD:α-AID construct behaved as a monomer in solution84, and the structure is very similar to that of an α-KD:α-AID from S. pombe86, where the arrangement of the α-AID and α-KD from the same molecule were very similar to that shown here. The view in (B) is of the same structure shown in Fig. 2, but only the α-KD and α-AID are shown.

The α-AID is connected to the α subunit C-terminal domain (α-CTD) by the α-linker, a region of extended polypeptide that wraps around one face of the γ subunit (Fig. 2) and is crucial in the mechanism for activation by AMP (discussed in more detail in Section 5.3). The α-CTD is a small globular domain that forms the interface with the C-terminal domain of the β subunit. An interesting feature of the α-CTD is that it ends in both the α1 and α2 isoforms with well-defined nuclear export sequences, although these have only been shown to be functional in the case of α288. Both isoforms also contain serine/threonine-rich sequences of about 50 residues that we term the ST loops89, which are discussed in Section 6 below.

5.2. The β subunits

When β subunit sequences are compared across isoforms and species, they contain two conserved regions, a central carbohydrate-binding module (β-CBM) and the C-terminal domain (β-CTD). The latter is a small compact domain that interacts with the α-CTD, and also contributes to an intrasubunit β-sheet containing two strands from the β-CTD and one from the N-terminus of γ. This architecture for assembling the three subunits is highly conserved throughout eukaryotes, from budding yeast90 to fission yeast91 and humans92. The β-CTD can be considered to form the core of the heterotrimeric AMPK complex, bridging the α and γ subunits.

The β-CBM is interesting because it is a member of the CBM48 family of carbohydrate-binding modules, non-catalytic domains usually found in enzymes that metabolize α1→6 linkages in carbohydrates, such as glycogen-branching enzymes and isoamylases93. The β-CBM causes a proportion of cellular AMPK to bind to glycogen particles94, 95, particularly in the case of the β2 isoform94 whose CBM appears to have a higher affinity for glycogen than that in β196. The carbohydrate-binding site is well defined, since crystal structures of isolated β-CBMs and a heterotrimeric α1β2γ1 complex (Fig. 2) have been solved in the presence of β-cyclodextrin, a circular heptasaccharide of α1→4-linked glucose units84, 97, 98. Until recently it had been unclear why only a proportion of AMPK in the cell is bound to glycogen, especially in skeletal muscle where β2 is the main β subunit isoform and where glycogen content can be very high. However, a recent paper shows that activated AMPK can autophosphorylate at Thr148 located within the β-CBM of β199, a residue known to be directly involved in the carbohydrate-binding site97. Phosphorylation at Thr148 prevents AMPK from binding to glycogen, although AMPK already bound to glycogen appears to be protected against autophosphorylation at this site99.

CBMs are present within the subunits of all eukaryotic AMPK orthologs, although higher plant orthologs contain unusual “βγ” subunits that contain a CBM fused at the N-terminus of a γ subunit, as well as more conventional β subunits with central CBMs100. The universal occurrence of CBMs within AMPK orthologs suggest that they have key physiological functions, although these remain incompletely understood. Since both the skeletal muscle (GYS1)101, 102 and liver (GYS2)103 isoforms of glycogen synthase are physiological targets that are inactivated after phosphorylation by AMPK, one function may be to co-localize AMPK with this glycogen-bound substrate. It has also been suggested that the β-CBM may allow AMPK to sense the structural state of glycogen and regulate glycogen synthesis according to the status of glycogen stores104, 105, although further work is required to confirm that hypothesis.

Despite the uncertain role of glycogen binding, another function of the β-CBM has become clear with exciting findings that a cleft between it and the N-lobe of the α-KD form the binding site for A-769662, 991 and salicylate65, 74. This cleft forms between the surface of the β-CBM opposite to the known carbohydrate-binding site, and the surface of the KD N-lobe opposite to the catalytic site. In the structure shown in Fig. 2, where the cleft was unoccupied, an electrostatic interaction between Lys29 and Lys31 from the N-lobe and the phosphate group on Ser108 of the β-CTD appeared to stabilize the interaction between the two domains84. In other structures, the side chain of Lys29 interacts with the carboxylate group at one end of 991, while the side chains of Lys29 and Lys31 are involved with the interaction with A-76966265, 74. These findings help to explain the requirement for autophosphorylation of Ser108 for full activation by A-76966275. Based on a crystal soaked with iodosalicylate, salicylates also appear to bind in this site, although the resolution was not sufficient to analyze the detailed molecular interactions74.

5.3. The γ subunits

The γ subunits contain at their C-terminal end four tandem repeats, termed cystathionine-beta-sythase 1 (CBS1) through CBS4, of a sequence motif of around 60 residues known as a CBS repeat. First recognized by bio-informatic analysis106, CBS repeats also occur in a small number of other proteins in the human genome. Most CBS-containing proteins have only two repeats that assemble into a structure known as a Bateman domain, with the cleft between the repeats often binding regulatory ligands containing adenosine, such as ATP or S-adenosyl methionine107. The γ subunits of AMPK and its orthologs are unusual in that they contain four repeats, thus generating two Bateman domains formed by CBS1/CBS2 and CBS3/CBS4 respectively. These assemble in a head-to-head manner to form a disc-like shape, with one CBS repeat in each quadrant of the disk; these are color-coded in Fig. 2, although much of CBS1, CBS2 and CBS3 are hidden in the view shown. This arrangement generates four pseudosymmetrical clefts in the center where ligands might bind, two accessible from one side of the disc and two from the other. Isolated γ subunits were originally reported to competitively bind just two molecules of AMP or ATP107, but when the core of the AMPK heterotrimer was crystallized in the presence of AMP, it was found to have three molecules of AMP bound in sites 1, 3 and 4 (the sites are numbered by convention according to the CBS repeat bearing an aspartate side chain that interacts with the ribose ring of the nucleotide; site 2 lacks an aspartate and appears to be unused). In the view shown in Fig. 2, part of a molecule of AMP is just visible in site 3, while sites 1 and 4 are hidden around the back of the γ subunit. Soaking of ATP into crystals made with AMP displaced AMP by ATP in sites 1 and 3, but not 4, leading to the idea that site 4 contains a permanently bound, “non-exchangeable” AMP92 and perhaps explaining why only two sites were detected in the original binding studies107. However, when another group crystallized the core complex with ATP (as opposed to soaking ATP into crystals made with AMP), they found that ATP was bound at sites 1 and 4, while site 3 was empty108.

The extended α-linker that connects the AID to the α-CTD (see Section 5.1) can be seen from the viewpoint of Fig. 2 to wrap around the front face of the γ subunit. One conserved region within this linker termed α-regulatory subunit interacting motif-1 (α-RIM1) interacts with the unused site 2, while another (α-RIM2) interacts with site 3. A highly conserved glutamate in α-RIM2 (Glu364 in human α1) interacts with Arg70 and Lys170 in γ1, which in turn interact with the phosphate group of AMP bound in site 3. This AMP- and site 3-dependent interaction between the γ subunit and the α-linker is proposed to cause the AID to move away from its inhibitory position behind the N-lobe of the kinase domain (Fig. 3A) into the position shown in Fig. 3B, thus explaining allosteric activation by AMP. If binding of ATP at site 3 did not allow the interaction with α-RIM2, this would also explain how ATP antagonizes activation by AMP. A variety of evidence now strongly supports this model:

  • (1)

    Mutations in both α-RIM1 and α-RIM2 expected to reduce interaction with the γ subunit (including mutation of Glu364), or their replacement by a shorter artificial linker, abolished allosteric activation by AMP74, 109.

  • (2)

    Singlet oxygen-mediated luminescence energy transfer (AlphaScreen) assays, which can monitor changes in the distance between donor and acceptor probes, were used to analyze interactions between a core α1β2γ1 heterotrimer (consisting of just the α- and β-CTDs and full length γ1) and a construct containing the AID, α-RIM1 and α-RIM2 from α1. Addition of AMP increased the interaction, whereas ATP decreased it.

  • (3)

    As mentioned in Section 4.2, the AMP analog C2 is rather selective for α1 complexes, with which it causes both allosteric activation and protection against dephosphorylation of Thr172. However, both effects of C2 could be transferred to α2 complexes merely by replacing α-RIM2 and the remainder of the α-linker from α2 with the equivalent region from α1. These results emphasize the importance of the α-linker in the dual mechanisms of activation by this AMP analog.

This model was also supported by AlphaScreen assays in which the donor and acceptor probes were attached to the N-termini of the α and γ subunits in a complete heterotrimer84. Addition of AMP yielded changes indicating that the probes moved together, suggesting the formation of a more compact conformation for the heterotrimer in the presence of AMP, as suggested by previous results obtained by small angle X-ray scattering in solution110. On the other hand, addition of ATP caused the probes to move apart, indicating a less compact conformation. This is consistent with the idea that the α-linker dissociates from the γ subunit in the inactive conformation in the presence of ATP, allowing the whole heterotrimer to adopt a more extended structure in which the AID interacts with and inhibits the kinase domain. These AlphaScreen assays also allowed the concentration dependence of the effects of AMP and ATP on these conformational changes to be measured, independently of their binding at the catalytic site. The results showed that the half-maximal effect (EC50) for the effect of AMP (measured in the absence of ATP) occurred at 0.95 µmol/L, whereas the EC50 for the effect of ATP was at 0.85 mmol/L, almost 1000-fold higher84. For comparison (although it is not possible to measure allosteric activation in the absence of ATP) the estimated EC50 values for allosteric activation of γ1 complexes by AMP in the presence of 0.2, 1 and 5 mmol/L ATP were 5.3, 22 and 140 µmol/L, showing that increasing concentrations of ATP compete with AMP at the γ subunit sites36.

5.4. Remaining challenges in understanding regulation by adenine nucleotides

Although the various crystal structures obtained over the last few years have yielded considerable insight into the mechanism of regulation by adenine nucleotides, several questions remain. One is the role of binding of AMP and other nucleotides at sites 1 and 4, especially given the evidence discussed in Section 5.3 that binding at site 3 recruits the α-RIM1 motif, which is crucial both for allosteric activation and for protection against dephosphorylation. Interestingly, mutation to alanine of any one of the three aspartate residues that bind the ribose rings of nucleotides at sites 1, 3 and 4 abolishes both allosteric activation and promotion of Thr172 phosphorylation111. The three nucleotide binding sites are located close together at the center of the γ subunit, and side chains of highly conserved basic residues from the γ subunit interact with phosphate groups of nucleotides in more than one site. For example, the side chains of His151 and His298 in human γ1 interact with phosphate groups of AMP in both sites 1 and 492. It therefore seems very likely that binding of nucleotides at these three sites will show mutual dependencies on each other, either positive or negative. Along these lines, the group who crystallized the core complex in the presence of ATP suggested that the mode of binding of ATP at site 4 would preclude binding of AMP (or any other nucleotide) at site 3. Thus, AMP may have to be bound at site 4 (and possibly also at site 1) before it binds at the crucial site 3.

Another question not full answered is how binding of AMP inhibits Thr172 dephosphorylation. Since the lack of ability of the AMP analog C2 to protect against dephosphorylation of α2 complexes can be restored by replacing α-RIM2 of α2 (which binds site 3 when AMP is bound) with the equivalent region from α1, it appears that it is binding of AMP at site 3 that is crucial for the effect. However, unlike allosteric activation by AMP, which does not require the presence of the β-CBM94, protection against Thr172 dephosphorylation by AMP does require it65, although the reasons for this are poorly understood.

Another puzzle is why ADP binding should provide protection against dephosphorylation of Thr17238 yet does not, like AMP binding, cause allosteric activation. This would be hard to explain if the effects of ADP and AMP were due to binding at the same site. However, studies of the budding yeast ortholog of AMPK suggest that the γ subunit SNF4 is not required for the response to glucose starvation112, and that binding of ADP to the catalytic site on the kinase domain, rather than to the γ subunit, may be responsible for its ability to protect against dephosphorylation of the site equivalent to Thr172113. In the same study, it was reported that binding of the kinase inhibitor staurosporine (which binds at the catalytic site38) to either the budding yeast or mammalian kinases provides protection against Thr172 dephosphorylation. Thus, it is possible that AMP and ADP protect against dephosphorylation by binding at different sites.

A final question that has not yet been illuminated by the structural studies concerns how phosphorylation of Thr172 by LKB1, but not CaMKKβ, is promoted by binding of AMP36. A radical proposal to explain this, which has been developed by Lin and colleagues114, 115 at Xiamen University, is that AMP binding to AMPK causes it to co-localize with LKB1 due to their mutual interactions with the scaffold protein axin, which in turn binds to late endosomal/lysosomal adaptor and MAPK and mTOR activator (LAMTOR1) at the surface of the lysosome. However, promotion by AMP of Thr172 phosphorylation by LKB1 can be observed on reconstitution of highly purified LKB1 and AMPK36, suggesting that the effect does not strictly require any of these additional components.

6. Non-canonical regulation by phosphorylation of the ST loop and other sites

The hormone insulin represents a signal that nutrients are available, with those nutrients (glucose, amino acids and fats) either directly triggering insulin release from the β cells of the pancreas, or doing so indirectly via release of incretins such as glucagon-like peptide-1 from the small intestine. Insulin then stimulates target cells to take up these nutrients and convert them to their storage forms of glycogen, triglycerides and proteins. Insulin-like growth factor-1 (IGF1), which acts via a signaling pathway closely related to that of insulin, is a growth factor that promotes biosynthesis and hence cell growth. Since AMPK is generally switched on under the opposite circumstances to insulin and IGF1 (lack of nutrients or energy) it is not surprising that the AKT/PKB (protein kinase B) pathway, the principal signaling pathway downstream of insulin and IGF1, should antagonize the AMPK pathway. In 2006 it was reported that AKT phosphorylated rat AMPK-α1 at Ser485 (equivalent to Ser487 in humans, with human numbering being used below, with the exception of Thr172). Evidence was presented that prior phosphorylation at Ser487 by AKT reduced subsequent phosphorylation at Thr172 and consequent activation by LKB1, and that this mechanism explained how prior treatment of perfused rat heart with insulin reduced AMPK activation during subsequent ischemia116.

Ser487 occurs within a region of around 50−55 residues in the AMPK-α subunits that we now term the “ST loop”. This is a serine/threonine rich region that is present in α-CTDs in all vertebrates and nematodes, but not in orthologs from insects, plants, fungi or protozoa. In all crystal structures of mammalian complexes containing an α-CTD, the ST loop was either not resolved, suggesting that it is disordered within the crystals (perhaps because it is not phosphorylated during bacterial expression), or had been replaced by a short artificial spacer in the construct crystallized, because it was thought that it might hinder crystallization. In these structures the ST loop therefore appears as a gap between the end of penultimate β-strand and the start of the last α-helix in the α-CTD. My group89 has recently confirmed that AKT efficiently phosphorylates Ser487 on AMPK-α1, although the equivalent residue on AMPK-α2, Ser491, is an extremely poor substrate for AKT—it is therefore important not to simply assume that the regulation of α1 and α2 by phosphorylation in this region will be identical. In fact, Ser491 on α2 is efficiently autophosphorylated by AMPK itself, and becomes phosphorylated in intact cells when AMPK, rather than AKT, is activated. By generating HEK-293 cells expressing wild type or mutant α1, we showed that prior activation of AKT using IGF1 inhibited subsequent Thr172 phosphorylation and AMPK-α1 activation in response to A-769662, and that this was blocked by a specific AKT inhibitor or by mutation of Ser487 to alanine. We also showed that the effect of Ser487 phosphorylation by AKT to inhibit subsequent phosphorylation at Thr172 on AMPK-α1 was identical using either LKB1 or CaMKKβ as the upstream kinase, suggesting that the mechanism may involve a simple physical occlusion of Thr172. Consistent with this, mutation of three basic residues in the α-C helix of the N-lobe, which are conserved in all vertebrate AMPK-α subunits but not in closely related kinases, abolished the inhibitory effect of AKT even though Ser487 was still phosphorylated. This suggested that the ST loop interacts with the α-C helix following its phosphorylation, thus reducing access to Thr17289.

ST loops also appear to be phosphorylated by other kinases. Hurley et al.117 reported that Ser487/491 on AMPK-α1 or -α2 (isoform not specified) was phosphorylated in response to cyclic AMP elevation in INS1 cells, a pancreatic β cell line, while a recombinant AMPK-α1 peptide was phosphorylated in cell-free assays at Ser487 by cyclic AMP-dependent protein kinase (PKA). Complicating this story, however, the effects of cyclic AMP-elevation were abolished in CaMKKβ-null mouse embryo fibroblasts, and CaMKKβ was inactivated by cyclic AMP-elevating agents, suggesting that effects in intact cells were mediated by modulation of CaMKKβ, rather than AMPK117. Using a bacterially expressed α1β1γ1 complex, PKA has been reported to phosphorylate not only Ser487 but also Ser499 and Ser175, and it was proposed that this limited AMPK activation, and hence inhibition of lipolysis, when PKA was activated in white adipocytes118. Like Ser487, Ser499 is located in the ST loop, but Ser175 is immediately adjacent to Thr174, the residue equivalent to Thr172 in human α1. Based on analysis of various mutations, the authors118 suggested that it was phosphorylation at Ser175 rather than Ser487 or Ser499 that blocked subsequent AMPK activation. A puzzling feature is why they did not observe any effects on subsequent Thr172 phosphorylation when Ser487 was phosphorylated by PKA, even though two other groups89, 116 have shown that there is a marked effect when Ser487 is phosphorylated by AKT. Finally, it has been reported that two residues in the ST loop just upstream of Ser487, i.e., Thr481 and Ser477, are phosphorylated by glycogen synthase kinase 3 (GSK3) when Ser487 has been phosphorylated119. GSK3 often phosphorylates serine or threonine side chains 4 residues N-terminal to a “priming” phosphoamino acid, although the spacing between Ser487 and Thr481 is six rather than four residues. It was proposed that phosphorylation of Ser477 and Thr481 inhibited net Thr172 phosphorylation by promoting its dephosphorylation. While these observations are interesting, the physiological rationale underlying inhibition of AMPK by GSK3 is difficult to grasp, because both GSK3 isoforms (α and β) are inactivated by phosphorylation by AKT, and because GSK3 usually acts to inhibit rather than promote anabolic pathways, similar to AMPK but opposite to AKT.

7. Regulation of AMPK by natural products used in traditional medicines

As mentioned in Section 4.1, over the last few years more than 100 different natural products have been shown to activate AMPK; a list of these, which is almost certainly not comprehensive, is shown in Table 141, 44, 49, 50, 59, 60, 69, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244, 245, 246, 247, 248, 249. Although many of them can be classed as polyphenols, their structures are very varied. The majority are products of plants used in herbal remedies, particularly in traditional Asian medicine. The mechanism by which most of them activate AMPK is unknown, and a puzzling feature is why so many natural plant products should all be AMPK activators. One clue is that among the small number of these activators where the mechanism has been established (given at the top of the list in Table 1), most are inhibitors of mitochondrial ATP synthesis, either by inhibiting Complex I of the respiratory chain, or by inhibiting the ATP synthase (Complex V). Most of the natural plant products that activate AMPK appear to be secondary metabolites, i.e., they are not required for plant growth, development or reproduction, and a reasonable working hypothesis is that many of them are molecules produced by plants to deter infection by pathogens, or grazing by insect or other herbivorous animals, to whom these molecules are toxic. In support of this idea, resveratrol is known to be produced by grapes in response to fungal infection250, while Galega officinalis, the source of galegine from which metformin and phenformin were derived, is classified as a noxious weed in the USA because it is poisonous to herbivorous animals (reflected in one of the common names for Galega officinalis, Goat’s Rue).

Table 1.

Partial list of natural products (mostly from plants) that have been reported to activate AMPK in intact cells or in vivo. Although a single source species is usually listed, most of the compounds are probably also produced by related species. The author compiled this list but has not read all of the papers cited as thoroughly as other papers discussed in this review. ?, unkown.

Natural product Source Mechanism Ref.
Antimycin A Streptomyces (bacteria) Inhibits Complex III 44
Apoptolidins A/C Nocardiopsis spp. (bacteria) Inhibits ATP synthase 120
Arctigenin Arctium lappa Inhibits Complex I 49, 121
Berberine Berberis spp., other plants Inhibits Complex I 41, 122
Cordycepin (3′-deoxyadenosine) Cordyceps militaris (fungus) Converted to AMP analog? 59, 60, 123
Galegine Galega officinalis Inhibits Complex I 50
Oligomycin Streptomyces (bacteria) Inhibits F1 ATP synthase 41
Quercetin Many plants Inhibits Complex I 124
Resveratrol Grapes, red wine Inhibits ATP synthase 41, 125, 126
Salicylate Salix alba (willow), other plants Binds to A-769662 site 69
Alternol Alternaria alternata ? 127
Anthocyanin fraction Purple sweet potato ? 128
Anthocyanin fraction Korean black bean ? 129
Apigenin Matricaria chamomilla ? 130
Artemisinin Artemisia annua ? 131
Aspalathin Aspalathus linearis ? 132
Bavachalcone Psoralea corylifolia ? 133
Caffeic acid All plants ? 134
Caffeic acid, phenethyl and phenylpropyl esters All plants ? 135
Celastrol Many plants ? 136
Chalcones Various plants ? 137
Chitosan Crustaceans ? 138
Chrysin Passiflora caerulea ? 139
Cucurbitane triterpenoids Siraitia grosvenorii ? 140
Curcumin Curcuma longa ? 141, 142
Cyanidin Daucus carota (black carrot) ? 143
Dehydrozingerone Zingiber officinale (ginger) ? 144
Delphinidin-3-glucoside Many plants ? 145
14-Deoxyandrographolide Andrographis paniculata ? 146
Dihydromyricetin Ampelopsis grossedentata ? 147
2-(2,4-Dihydroxyphenyl)-5-(E)-propenylbenzofuran Krameria lappacea ? 148
Emodin Rheum emodi ? 149, 150, 151
ENERGI-F704 Bamboo ? 152, 153
Epigallocatechin gallate Camellia sinensis ? 124, 154
Ergostatrien-3β-ol Antrodia camphorata ? 155
Eugenol Clove oil, nutmeg, cinnamon, basil ? 156
Fargesin Magnolia spp. ? 157
Foenumoside B Lysimachia foenum-graecum ? 158
Fucoidan Brown seaweeds ? 159
Fungal extract Clitocybe nuda ? 160, 161
Gallic acid Many plants ? 162
Geraniol Rose/palmarosa/citronella oils ? 163
GGEx18 Traditional Korean medicine ? 164
6-Gingerol Zingiber officinale (ginger) ? 165, 166, 167
Ginsenosides Panax ginseng ? 168−172
Glabridin Glycyrrhiza glabra ? 173, 174
Green tea extract Camellia sinensis ? 175, 176
Hispidulin Saussurea involucrate ? 177, 178, 179
Honokiol Magnolia grandiflora ? 180, 181
Hugan Qingzhi tablet Chinese herbal medicine ? 182
Indazole-type alkaloids Nigella sativa ? 183
Isoquercitrin Many plants ? 184
Isorhamnetin Tagetes lucida ? 185
Jinlida granule Chinese herbal medicine ? 186
Jinqi formula Coptidis rhizome/Astragali rhadix/Lonicerae japonicae ? 187
Karanjin Pongamia pinnata ? 188
Kazinol C Broussonetia kazinoki ? 189
Licochalcone Glycyrrhiza glabra (licorice) ? 190
Lindenenyl acetate Lindera strychnifolia ? 191
Luteolin Many plants ? 124
Malvidin Daucus carota (black carrot) ? 143
Mangiferin Iris unguicularis ? 192, 193, 194, 195
Methyl cinnamate Zanthoxylum armatum ? 196
4-O-methylhonokiol analog Magnolia grandiflora ? 197
2-Methyl-7-hydroxymethyl-1,4-naphthoquinone Pyrola rotundifolia ? 198
Monascin/ankaflavin Monascus pilosus (a fungus) ? 199
Monascuspiloin Monascus pilosus (a fungus) ? 200
Naringin Citrus x paradisi ? 201
Nectrandin B Myristica fragrans (nutmeg) ? 202, 203, 204
Octaphlorethol A Ishige foliacea (a brown alga) ? 205
Oleanolic acid Many plants ? 206
Osthole Cnidium monnieri ? 207, 208
Parthenolide Tanacetum parthenium (feverfew) ? 209
Persimmon tannin Diospyros kaki (persimmon) ? 210
Petasin Petasites spp. ? 211
Piperlongumine Piper longum ? 212
Plant extract Boesenbergia pandurata ? 213
Plant extract Cirsium japonicum ? 193
Plant extract Houttuynia cordata ? 214
Plant extract Impatiens balsamina ? 215
Plant extract Lycium barbarum ? 216
Plant extract Malva verticillata ? 217
Plant extract Remotiflori radix ? 218
Plant extract Rhus verniciflua Stokes ? 219
Plant extract Scutellaria baicalensis ? 220
Plant extract Sechium edule ? 221
Plant extract Taraxacum mongolicum ? 222
Plant extract Theobroma cacao (cocoa) ? 223
Plant extract Viola mandshurica ? 224
Plant extract Vitis thunbergii ? 225
Pomolic acid Chrysobalanus icaco ? 226
Pterostilbene Grapes, other fruits ? 227
Puerarin Radix puerariae ? 228
ReishiMax Ganoderma lucidum ? 229
Rhizochalin (aglycone) Rhizochalina incrustata (a sponge) ? 230
S-methylmethionine sulfonium chloride Many plants ? 231
Salidroside Rhodiola rosea ? 232
Saponins Rubus parvifolius ? 233
Scopoletin Scopolia spp. ? 234
Soybean peptides Glycine max (soybean) ? 235
Sulforaphane Brassica oleracea ? 236
Tangeretin Citrus tangerine (tangerine) ? 237
Tanshinone IIA Salvia miltiorrhiza ? 238, 239
Theaflavins Camellia sinensis (tea) ? 240
Theasinsensins Camellia sinensis (tea) ? 241
Thymoquinone Nigella sativa ? 242
Tiliroside Rose hips, strawberry, raspberry ? 243
Tormentic acid Eriobotrya japonica ? 244
Trans-cinnamic acid Cinnamon ? 245
Triterpenoid saponins Stauntonia chinensis ? 246
Ursolic acid Mirabilis jalapa, other plants ? 247, 248
Xanthigen Punica granatum ? 249

Why should plants produce inhibitors of mitochondrial function as defensive chemicals? The respiratory chain and the ATP synthase contain five large hydrophobic multiprotein complexes, with Complex I containing no less than 44 protein subunits, while the ATP synthase has at least 14. It seems probable that many different hydrophobic, xenobiotic compounds might find a binding site in one or more of these complexes that would inhibit their function. Many secondary metabolites of plants are stored in the cell vacuole (equivalent to the lysosome of animal cells), and are therefore kept away from their own mitochondria. The production of mitochondrial poisons might therefore be a useful general approach for plants to produce compounds that would deter infection or grazing. However, in line with the aphorism of Paracelsus that “the dose makes the poison”, lower doses of these compounds that are not sufficient to fully inhibit mitochondrial function might still have useful therapeutic effects by activating AMPK.

It is also interesting to note that the barbiturate drug, phenobarbital, activates AMPK in an AMP-dependent manner by inhibiting the respiratory chain41. In hepatocytes, AMPK activation is required for phenobarbital to induce expression of genes (e.g., CYP2B6) encoding enzymes of the cytochrome P450 (CYP) family, via the constitutively active/androstane receptor, constitutive active/androstane receptor (CAR)251, 252. Some classes of CYP enzymes (especially the CYP1/CYP2/CYP3 families) catalyze the initial steps in metabolism of drugs and other hydrophobic xenobiotics, making them more soluble for excretion. Plant products that are defensive agents inhibiting mitochondrial ATP synthesis would activate AMPK, and induction of CYP enzymes by AMPK might then be a good general way for the animal to mount a response to deal with potential poisoning by these xenobiotics.

8. Conclusions and perspectives

Most indications for drugs targeting AMPK suggest that activators rather than inhibitors would be therapeutically beneficial. In general, development of activators is probably more difficult than development of inhibitors, but the fact that there are already many known activators of AMPK, acting by three or four different mechanisms, shows that this goal is reachable. Many of the activators already known are natural plant products, or derivatives of natural products, that originate from traditional medicines. Two of these, metformin and salicylate, are already among the most successful and widely used drugs of all time, although the extent to which their therapeutic effects are mediated by AMPK is still being debated. Of the many natural plant products whose mechanism of activation of AMPK has not yet been elucidated, my suspicion is that most of them will turn out to be compounds used by plants for defensive purposes, most of which are likely to activate AMPK indirectly by inhibiting mitochondrial ATP synthesis. In such cases, the question must always be asked whether the new agent is more effective than metformin, and whether it has fewer side effects. However, there may also be some direct activators among the long list of natural products in Table 1, and this is certainly an avenue worth pursuing. Of the known binding sites on AMPK where ligand binding can cause activation, the A-769662/salicylate-binding site is perhaps the easiest to target for drug development, although the AMP-binding sites can also be targeted by pro-drugs such as AICAR or C13. It will be fascinating to see whether the current effort to develop novel AMPK activators will result in any clinically useful drugs over the next few years.

Acknowledgments

Grahame Hardie is funded by a Senior Investigator Award (No. 097726) from the Wellcome Trust and a programme grant (No. C37030/A15101) from Cancer Research UK. I would like to thank all of the current members of my laboratory (Romana Auciello, Alex Gray, Fiona Ross, Fiona Russell, Graeme Gowans, Simon Hawley and Diana Vara-Ciruelos) for discussions that have helped in putting this review together.

Footnotes

Peer review under responsibility of Institute of Materia Medica, Chinese Academy of Medical Sciences and Chinese Pharmaceutical Association.

References

  • 1.Hardie D.G. AMPK-sensing energy while talking to other signaling pathways. Cell Metab. 2014;20:939–952. doi: 10.1016/j.cmet.2014.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Hardie D.G. AMP-activated protein kinase: maintaining energy homeostasis at the cellular and whole-body levels. Annu Rev Nutr. 2014;34:31–55. doi: 10.1146/annurev-nutr-071812-161148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Hardie D.G. AMPK: positive and negative regulation, and its role in whole-body energy homeostasis. Curr Opin Cell Biol. 2014;33:1–7. doi: 10.1016/j.ceb.2014.09.004. [DOI] [PubMed] [Google Scholar]
  • 4.Hardie D.G. AMP-activated protein kinase: a key regulator of energy balance with many roles in human disease. J Intern Med. 2014;276:543–559. doi: 10.1111/joim.12268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.O’Neill L.A., Hardie D.G. Metabolism of inflammation limited by AMPK and pseudo-starvation. Nature. 2013;493:346–355. doi: 10.1038/nature11862. [DOI] [PubMed] [Google Scholar]
  • 6.Sim A.T.R., Hardie D.G. The low activity of acetyl-CoA carboxylase in basal and glucagon-stimulated hepatocytes is due to phosphorylation by the AMP-activated protein kinase and not cyclic AMP-dependent protein kinase. FEBS Lett. 1988;233:294–298. doi: 10.1016/0014-5793(88)80445-9. [DOI] [PubMed] [Google Scholar]
  • 7.Munday M.R., Campbell D.G., Carling D., Hardie D.G. Identification by amino acid sequencing of three major regulatory phosphorylation sites on rat acetyl-CoA carboxylase. Eur J Biochem. 1988;175:331–338. doi: 10.1111/j.1432-1033.1988.tb14201.x. [DOI] [PubMed] [Google Scholar]
  • 8.Carling D., Zammit V.A., Hardie D.G. A common bicyclic protein kinase cascade inactivates the regulatory enzymes of fatty acid and cholesterol biosynthesis. FEBS Lett. 1987;223:217–222. doi: 10.1016/0014-5793(87)80292-2. [DOI] [PubMed] [Google Scholar]
  • 9.Birk J.B., Wojtaszewski JFP. Predominant α1/β2/γ3 AMPK activation during exercise in human skeletal muscle. J Physiol. 2006;577:1021–1032. doi: 10.1113/jphysiol.2006.120972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Katinka M.D., Duprat S., Cornillot E., Méténier G., Thomarat F., Prensier G. Genome sequence and gene compaction of the eukaryote parasite Encephalitozoon cuniculi. Nature. 2001;414:450–453. doi: 10.1038/35106579. [DOI] [PubMed] [Google Scholar]
  • 11.Miranda-Saavedra D., Stark M.J.R., Packer J.C., Vivares C.P., Doerig C., Barton G.J. The complement of protein kinases of the microsporidium Encephalitozoon cuniculi in relation to those of Saccharomyces cerevisiae and Schizosaccharomyces pombe. BMC Genomics. 2007;8:309. doi: 10.1186/1471-2164-8-309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Williams B.A.P., Cali A., Takvorian P.M., Keeling P.J. Distinct localization patterns of two putative mitochondrial proteins in the microsporidian Encephalitozoon cuniculi. J Eukaryot Microbiol. 2008;55:131–133. doi: 10.1111/j.1550-7408.2008.00315.x. [DOI] [PubMed] [Google Scholar]
  • 13.Tsaousis A.D., Kunji E.R.S., Goldberg A.V., Lucocq J.M., Hirt R.P., Embley T.M. A novel route for ATP acquisition by the remnant mitochondria of Encephalitozoon cuniculi. Nature. 2008;453:553–556. doi: 10.1038/nature06903. [DOI] [PubMed] [Google Scholar]
  • 14.Haurie V., Boucherie H., Sagliocco F. The SNF1 protein kinase controls the induction of genes of the iron uptake pathway at the diauxic shift in Saccharomyces cerevisiae. J Biol Chem. 2003;278:45391–45396. doi: 10.1074/jbc.M307447200. [DOI] [PubMed] [Google Scholar]
  • 15.Zong H.H., Ren J.M., Young L.H., Pypaert M., Mu J., Birnbaum M.J. AMP kinase is required for mitochondrial biogenesis in skeletal muscle in response to chronic energy deprivation. Proc Natl Acad Sci U S A. 2002;99:15983–15987. doi: 10.1073/pnas.252625599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Jäger S., Handschin C., St-Pierre J., Spiegelman B.M. AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1α. Proc Natl Acad Sci U S A. 2007;104:12017–12022. doi: 10.1073/pnas.0705070104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Cantó C., Jiang L.Q., Deshmukh A.S., Mataki C., Coste A., Lagouge M. Interdependence of AMPK and SIRT1 for metabolic adaptation to fasting and exercise in skeletal muscle. Cell Metab. 2010;11:213–219. doi: 10.1016/j.cmet.2010.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hardie D.G., Hawley S.A. AMP-activated protein kinase: the energy charge hypothesis revisited. Bioessays. 2001;23:1112–1119. doi: 10.1002/bies.10009. [DOI] [PubMed] [Google Scholar]
  • 19.Hardie D.G., Ross F.A., Hawley S.A. AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nat Rev Mol Cell Biol. 2012;13:251–262. doi: 10.1038/nrm3311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Hawley S.A., Davison M., Woods A., Davies S.P., Beri R.K., Carling D. Characterization of the AMP-activated protein kinase kinase from rat liver, and identification of threonine 172 as the major site at which it phosphorylates and activates AMP-activated protein kinase. J Biol Chem. 1996;271:27879–27887. doi: 10.1074/jbc.271.44.27879. [DOI] [PubMed] [Google Scholar]
  • 21.Hawley S.A., Boudeau J., Reid J.L., Mustard K.J., Udd L., Mäkelä T.P. et al. Complexes between the LKB1 tumor suppressor, STRADα/β and MO25α/β are upstream kinases in the AMP-activated protein kinase cascade. J Biol. 2003;2:28. doi: 10.1186/1475-4924-2-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Woods A., Johnstone S.R., Dickerson K., Leiper F.C., Fryer L.G., Neumann D. LKB1 is the upstream kinase in the AMP-activated protein kinase cascade. Curr Biol. 2003;13:2004–2008. doi: 10.1016/j.cub.2003.10.031. [DOI] [PubMed] [Google Scholar]
  • 23.Shaw R.J., Kosmatka M., Bardeesy N., Hurley R.L., Witters L.A., DePinho R.A. The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc Natl Acad Sci U S A. 2004;101:3329–3335. doi: 10.1073/pnas.0308061100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sakamoto K., Göransson O., Hardie D.G., Alessi D.R. Activity of LKB1 and AMPK-related kinases in skeletal muscle: effects of contraction, phenformin, and AICAR. Am J Physiol Endocrinol Metab. 2004;287:E310–E317. doi: 10.1152/ajpendo.00074.2004. [DOI] [PubMed] [Google Scholar]
  • 25.Sakamoto K., McCarthy A., Smith D., Green K.A., Hardie D.G., Ashworth A. Deficiency of LKB1 in skeletal muscle prevents AMPK activation and glucose uptake during contraction. EMBO J. 2005;24:1810–1820. doi: 10.1038/sj.emboj.7600667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hawley S.A., Pan D.A., Mustard K.J., Ross L., Bain J., Edelman A.M. Calmodulin-dependent protein kinase kinase-β is an alternative upstream kinase for AMP-activated protein kinase. Cell Metab. 2005;2:9–19. doi: 10.1016/j.cmet.2005.05.009. [DOI] [PubMed] [Google Scholar]
  • 27.Woods A., Dickerson K., Heath R., Hong S.P., Momcilovic M., Johnstone S.R. Ca2+/calmodulin-dependent protein kinase kinase-β acts upstream of AMP-activated protein kinase in mammalian cells. Cell Metab. 2005;2:21–33. doi: 10.1016/j.cmet.2005.06.005. [DOI] [PubMed] [Google Scholar]
  • 28.Hurley R.L., Anderson K.A., Franzone J.M., Kemp B.E., Means A.R., Witters L.A. The Ca2+/calmoldulin-dependent protein kinase kinases are AMP-activated protein kinase kinases. J Biol Chem. 2005;280:29060–29066. doi: 10.1074/jbc.M503824200. [DOI] [PubMed] [Google Scholar]
  • 29.Kishi K., Yuasa T., Minami A., Yamada M., Hagi A., Hayashi H. AMP-activated protein kinase is activated by the stimulations of Gq-coupled receptors. Biochem Biophys Res Commun. 2000;276:16–22. doi: 10.1006/bbrc.2000.3417. [DOI] [PubMed] [Google Scholar]
  • 30.Stahmann N., Woods A., Carling D., Heller R. Thrombin activates AMP-activated protein kinase in endothelial cells via a pathway involving Ca2+/calmodulin-dependent protein kinase kinase β. Mol Cell Biol. 2006;26:5933–5945. doi: 10.1128/MCB.00383-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Yang Y.L., Atasoy D., Su H.H., Sternson S.M. Hunger states switch a flip-flop memory circuit via a synaptic AMPK-dependent positive feedback loop. Cell. 2011;146:992–1003. doi: 10.1016/j.cell.2011.07.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Momcilovic M., Hong S.P., Carlson M. Mammalian TAK1 activates SNF1 protein kinase in yeast and phosphorylates AMP-activated protein kinase in vitro. J Biol Chem. 2006;281:25336–25343. doi: 10.1074/jbc.M604399200. [DOI] [PubMed] [Google Scholar]
  • 33.Herrero-Martín G., Høyer-Hansen M., García-García C., Fumarola C., Farkas T., López-Rivas A. TAK1 activates AMPK-dependent cytoprotective autophagy in TRAIL-treated epithelial cells. EMBO J. 2009;28:677–685. doi: 10.1038/emboj.2009.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Yeh L.A., Lee K.H., Kim K.H. Regulation of rat liver acetyl-CoA carboxylase. Regulation of phosphorylation and inactivation of acetyl-CoA carboxylase by the adenylate energy charge. J Biol Chem. 1980;255:2308–2314. [PubMed] [Google Scholar]
  • 35.Moore F., Weekes J., Hardie D.G. Evidence that AMP triggers phosphorylation as well as direct allosteric activation of rat liver AMP-activated protein kinase. A sensitive mechanism to protect the cell against ATP depletion. Eur J Biochem. 1991;199:691–697. doi: 10.1111/j.1432-1033.1991.tb16172.x. [DOI] [PubMed] [Google Scholar]
  • 36.Gowans G.J., Hawley S.A., Ross F.A., Hardie D.G. AMP is a true physiological regulator of AMP-activated protein kinase by both allosteric activation and enhancing net phosphorylation. Cell Metab. 2013;18:556–566. doi: 10.1016/j.cmet.2013.08.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Oakhill J.S., Steel R., Chen Z.P., Scott J.W., Ling N.M., Tam S. AMPK is a direct adenylate charge-regulated protein kinase. Science. 2011;332:1433–1435. doi: 10.1126/science.1200094. [DOI] [PubMed] [Google Scholar]
  • 38.Xiao B., Sanders M.J., Underwood E., Heath R., Mayer F.V., Carmena D. Structure of mammalian AMPK and its regulation by ADP. Nature. 2011;472:230–233. doi: 10.1038/nature09932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Oakhill J.S., Scott J.W., Kemp B.E. AMPK functions as an adenylate charge-regulated protein kinase. Trends Endocrinol Metab. 2012;23:125–132. doi: 10.1016/j.tem.2011.12.006. [DOI] [PubMed] [Google Scholar]
  • 40.Winder W.W., Hardie D.G. AMP-activated protein kinase, a metabolic master switch: possible roles in type 2 diabetes. Am J Physiol. 1999;277:E1–10. doi: 10.1152/ajpendo.1999.277.1.E1. [DOI] [PubMed] [Google Scholar]
  • 41.Hawley S.A., Ross F.A., Chevtzoff C., Green K.A., Evans A., Fogarty S. Use of cells expressing γ subunit variants to identify diverse mechanisms of AMPK activation. Cell Metab. 2010;11:554–565. doi: 10.1016/j.cmet.2010.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Owen M.R., Doran E., Halestrap A.P. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem J. 2000;348:607–614. [PMC free article] [PubMed] [Google Scholar]
  • 43.El-Mir M.Y., Nogueira V., Fontaine E., Avéret N., Rigoulet M., Leverve X. Dimethylbiguanide inhibits cell respiration via an indirect effect targeted on the respiratory chain complex I. J Biol Chem. 2000;275:223–228. doi: 10.1074/jbc.275.1.223. [DOI] [PubMed] [Google Scholar]
  • 44.Witters L.A., Nordlund A.C., Marshall L. Regulation of intracellular acetyl-CoA carboxylase by ATP depletors mimics the action of the 5′-AMP-activated protein kinase. Biochem Biophys Res Commun. 1991;181:1486–1492. doi: 10.1016/0006-291x(91)92107-u. [DOI] [PubMed] [Google Scholar]
  • 45.Gledhill J.R., Montgomery M.G., Leslie A.G.W., Walker J.E. Mechanism of inhibition of bovine F1-ATPase by resveratrol and related polyphenols. Proc Natl Acad Sci U S A. 2007;104:13632–13637. doi: 10.1073/pnas.0706290104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Jensen T.E., Ross F.A., Kleinert M., Sylow L., Knudsen J.R., Gowans G.J. PT-1 selectively activates AMPK-γ1 complexes in mouse skeletal muscle, but activates all three γ subunit complexes in cultured human cells by inhibiting the respiratory chain. Biochem J. 2015;467:461–472. doi: 10.1042/BJ20141142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Pang T., Zhang Z.S., Gu M., Qiu B.Y., Yu L.F., Cao P.R. Small molecule antagonizes autoinhibition and activates AMP-activated protein kinase in cells. J Biol Chem. 2008;283:16051–16060. doi: 10.1074/jbc.M710114200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Turner N., Li J.Y., Gosby A., To S.W.C., Cheng Z., Miyoshi H. Berberine and its more biologically available derivative, dihydroberberine, inhibit mitochondrial respiratory complex I: a mechanism for the action of berberine to activate AMP-activated protein kinase and improve insulin action. Diabetes. 2008;57:1414–1418. doi: 10.2337/db07-1552. [DOI] [PubMed] [Google Scholar]
  • 49.Huang S.L., Yu R.T., Gong J., Feng Y., Dai Y.L., Hu F. Arctigenin, a natural compound, activates AMP-activated protein kinase via inhibition of mitochondria complex I and ameliorates metabolic disorders in ob/ob mice. Diabetologia. 2012;55:1469–1481. doi: 10.1007/s00125-011-2366-3. [DOI] [PubMed] [Google Scholar]
  • 50.Mooney M.H., Fogarty S., Stevenson C., Gallagher A.M., Palit P., Hawley S.A. Mechanisms underlying the metabolic actions of galegine that contribute to weight loss in mice. Br J Pharmacol. 2008;153:1669–1677. doi: 10.1038/bjp.2008.37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Jenkins Y., Sun T.Q., Markovtsov V., Foretz M., Li W., Nguyen H. AMPK activation through mitochondrial regulation results in increased substrate oxidation and improved metabolic parameters in models of diabetes. PLoS One. 2013;8:e81870. doi: 10.1371/journal.pone.0081870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Gadalla A.E., Pearson T., Currie A.J., Dale N., Hawley S.A., Sheehan M. AICA riboside both activates AMP-activated protein kinase and competes with adenosine for the nucleoside transporter in the CA1 region of the rat hippocampus. J Neurochem. 2004;88:1272–1282. doi: 10.1046/j.1471-4159.2003.02253.x. [DOI] [PubMed] [Google Scholar]
  • 53.Day P., Sharff A., Parra L., Cleasby A., Williams M., Hörer S. Structure of a CBS-domain pair from the regulatory γ1 subunit of human AMPK in complex with AMP and ZMP. Acta Crystallogr D Biol Crystallogr. 2007;63:587–596. doi: 10.1107/S0907444907009110. [DOI] [PubMed] [Google Scholar]
  • 54.Corton J.M., Gillespie J.G., Hawley S.A., Hardie D.G. 5-Aminoimidazole-4-carboxamide ribonucleoside: a specific method for activating AMP-activated protein kinase in intact cells? Eur J Biochem. 1995;229:558–565. doi: 10.1111/j.1432-1033.1995.tb20498.x. [DOI] [PubMed] [Google Scholar]
  • 55.Pirkmajer S., Kulkarni S.S., Tom R.Z., Ross F.A., Hawley S.A., Hardie D.G. Methotrexate promotes glucose uptake and lipid oxidation in skeletal muscle via AMPK activation. Diabetes. 2015;64:360–369. doi: 10.2337/db14-0508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Racanelli A.C., Rothbart S.B., Heyer C.L., Moran R.G. Therapeutics by cytotoxic metabolite accumulation: pemetrexed causes ZMP accumulation, AMPK activation, and mammalian target of rapamycin inhibition. Cancer Res. 2009;69:5467–5474. doi: 10.1158/0008-5472.CAN-08-4979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Gómez-Galeno J.E., Dang Q., Nguyen T.H., Boyer S.H., Grote M.P., Sun Z. A potent and selective AMPK activator that inhibits de novo lipogenesis. ACS Med Chem Lett. 2010;1:478–482. doi: 10.1021/ml100143q. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Hunter R.W., Foretz M., Bultot L., Fullerton M.D., Deak M., Ross F.A. Mechanism of action of compound-13: an α1-selective small molecule activator of AMPK. Chem Biol. 2014;21:866–879. doi: 10.1016/j.chembiol.2014.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Guo P., Kai Q., Gao J., Lian Z.Q., Wu C.M., Wu C.A. Cordycepin prevents hyperlipidemia in hamsters fed a high-fat diet via activation of AMP-activated protein kinase. J Pharmacol Sci. 2010;113:395–403. doi: 10.1254/jphs.10041fp. [DOI] [PubMed] [Google Scholar]
  • 60.Wu C.M., Guo Y.S., Su Y., Zhang X., Luan H., Zhang X.P. Cordycepin activates AMP-activated protein kinase (AMPK) via interaction with the γ1 subunit. J Cell Mol Med. 2014;18:293–304. doi: 10.1111/jcmm.12187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Cool B., Zinker B., Chiou W., Kifle L., Cao N., Perham M. Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes and the metabolic syndrome. Cell Metab. 2006;3:403–416. doi: 10.1016/j.cmet.2006.05.005. [DOI] [PubMed] [Google Scholar]
  • 62.Göransson O., McBride A., Hawley S.A., Ross F.A., Shpiro N., Foretz M. Mechanism of action of A-769662, a valuable tool for activation of AMP-activated protein kinase. J Biol Chem. 2007;282:32549–32560. doi: 10.1074/jbc.M706536200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Sanders M.J., Ali Z.S., Hegarty B.D., Heath R., Snowden M.A., Carling D. Defining the mechanism of activation of AMP-activated protein kinase by the small molecule A-769662, a member of the thienopyridone family. J Biol Chem. 2007;282:32539–32548. doi: 10.1074/jbc.M706543200. [DOI] [PubMed] [Google Scholar]
  • 64.Scott J.W., van Denderen B.J.W., Jorgensen S.B., Honeyman J.E., Steinberg G.R., Oakhill J.S. Thienopyridone drugs are selective activators of AMP-activated protein kinase β1-containing complexes. Chem Biol. 2008;15:1220–1230. doi: 10.1016/j.chembiol.2008.10.005. [DOI] [PubMed] [Google Scholar]
  • 65.Xiao B., Sanders M.J., Carmena D., Bright N.J., Haire L.F., Underwood E. Structural basis of AMPK regulation by small molecule activators. Nat Commun. 2013;4:3017. doi: 10.1038/ncomms4017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Lai Y.C., Kviklyte S., Vertommen D., Lantier L., Foretz M., Viollet B. A small-molecule benzimidazole derivative that potently activates AMPK to increase glucose transport in skeletal muscle: comparison with effects of contraction and other AMPK activators. Biochem J. 2014;460:363–375. doi: 10.1042/BJ20131673. [DOI] [PubMed] [Google Scholar]
  • 67.Zadra G., Photopoulos C., Tyekucheva S., Heidari P., Weng Q.P., Fedele G. A novel direct activator of AMPK inhibits prostate cancer growth by blocking lipogenesis. EMBO Mol Med. 2014;6:519–538. doi: 10.1002/emmm.201302734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Benziane B., Björnholm M., Lantier L., Viollet B., Zierath J.R., Chibalin A.V. AMP-activated protein kinase activator A-769662 is an inhibitor of the Na+-K+-ATPase. Am J Physiol Cell Physiol. 2009;297:C1554–C1566. doi: 10.1152/ajpcell.00010.2009. [DOI] [PubMed] [Google Scholar]
  • 69.Hawley S.A., Fullerton M.D., Ross F.A., Schertzer J.D., Chevtzoff C., Walker K.J. The ancient drug salicylate directly activates AMP-activated protein kinase. Science. 2012;336:918–922. doi: 10.1126/science.1215327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Jeffreys D. Bloomsbury Publishing; London: 2004. Aspirin: the remarkable story of a wonder drug. [Google Scholar]
  • 71.Vane J.R. Inhibition of prostaglandin synthesis as a mechanism of action for aspirin-like drugs. Nat New Biol. 1971;231:232–235. doi: 10.1038/newbio231232a0. [DOI] [PubMed] [Google Scholar]
  • 72.Vane J.R., Botting R.M. The mechanism of action of aspirin. Thromb Res. 2003;110:255–258. doi: 10.1016/s0049-3848(03)00379-7. [DOI] [PubMed] [Google Scholar]
  • 73.Amann R., Peskar B.A. Anti-inflammatory effects of aspirin and sodium salicylate. Eur J Pharmacol. 2002;447:1–9. doi: 10.1016/s0014-2999(02)01828-9. [DOI] [PubMed] [Google Scholar]
  • 74.Calabrese M.F., Rajamohan F., Harris M.S., Caspers N.L., Magyar R., Withka J.M. Structural basis for AMPK activation: natural and synthetic ligands regulate kinase activity from opposite poles by different molecular mechanisms. Structure. 2014;22:1161–1172. doi: 10.1016/j.str.2014.06.009. [DOI] [PubMed] [Google Scholar]
  • 75.Scott J.W., Ling N.M., Issa S.M.A., Dite T.A., O’Brien M.T., Chen Z.P. Small molecule drug A-769662 and AMP synergistically activate naive AMPK independent of upstream kinase signaling. Chem Biol. 2014;21:619–627. doi: 10.1016/j.chembiol.2014.03.006. [DOI] [PubMed] [Google Scholar]
  • 76.Choi S.L., Kim S.J., Lee K.T., Kim J., Mu J., Birnbaum M.J. The regulation of AMP-activated protein kinase by H2O2. Biochem Biophys Res Commun. 2001;287:92–97. doi: 10.1006/bbrc.2001.5544. [DOI] [PubMed] [Google Scholar]
  • 77.Zmijewski J.W., Banerjee S., Bae H., Friggeri A., Lazarowski E.R., Abraham E. Exposure to hydrogen peroxide induces oxidation and activation of AMP-activated protein kinase. J Biol Chem. 2010;285:33154–33164. doi: 10.1074/jbc.M110.143685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Auciello F.R., Ross F.A., Ikematsu N., Hardie D.G. Oxidative stress activates AMPK in cultured cells primarily by increasing cellular AMP and/or ADP. FEBS Lett. 2014;588:3361–3366. doi: 10.1016/j.febslet.2014.07.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Shao D., Oka S., Liu T., Zhai P.Y., Ago T., Sciarretta S. A redox-dependent mechanism for regulation of AMPK activation by thioredoxin1 during energy starvation. Cell Metab. 2014;19:232–245. doi: 10.1016/j.cmet.2013.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Wang D.S., Jonker J.W., Kato Y., Kusuhara H., Schinkel A.H., Sugiyama Y. Involvement of organic cation transporter 1 in hepatic and intestinal distribution of metformin. J Pharmacol Exp Ther. 2002;302:510–515. doi: 10.1124/jpet.102.034140. [DOI] [PubMed] [Google Scholar]
  • 81.Foretz M., Hébrard S., Leclerc J., Zarrinpashneh E., Soty M., Mithieux G. Metformin inhibits hepatic gluconeogenesis in mice independently of the LKB1/AMPK pathway via a decrease in hepatic energy state. J Clin Invest. 2010;120:2355–2369. doi: 10.1172/JCI40671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Fullerton M.D., Galic S., Marcinko K., Sikkema S., Pulinilkunnil T., Chen Z.P. Single phosphorylation sites in ACC1 and ACC2 regulate lipid homeostasis and the insulin-sensitizing effects of metformin. Nat Med. 2013;19:1649–1654. doi: 10.1038/nm.3372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Higgs G.A., Salmon J.A., Henderson B., Vane J.R. Pharmacokinetics of aspirin and salicylate in relation to inhibition of arachidonate cyclooxygenase and antiinflammatory activity. Proc Natl Acad Sci U S A. 1987;84:1417–1420. doi: 10.1073/pnas.84.5.1417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Li X.D., Wang L.L., Zhou X.E., Ke J.Y., de Waal P.W., Gu X. Structural basis of AMPK regulation by adenine nucleotides and glycogen. Cell Res. 2015;25:50–66. doi: 10.1038/cr.2014.150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Pang T., Xiong B., Li J.Y., Qiu B.Y., Jin G.Z., Shen J.K. Conserved α-helix acts as autoinhibitory sequence in AMP-activated protein kinase α subunits. J Biol Chem. 2007;282:495–506. doi: 10.1074/jbc.M605790200. [DOI] [PubMed] [Google Scholar]
  • 86.Chen L., Jiao Z.H., Zheng L.S., Zhang Y.Y., Xie S.T., Wang Z.X. Structural insight into the autoinhibition mechanism of AMP-activated protein kinase. Nature. 2009;459:1146–1149. doi: 10.1038/nature08075. [DOI] [PubMed] [Google Scholar]
  • 87.Taylor S.S., Kornev A.P. Protein kinases: evolution of dynamic regulatory proteins. Trends Biochem Sci. 2011;36:65–77. doi: 10.1016/j.tibs.2010.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kazgan N., Williams T., Forsberg L.J., Brenman J.E. Identification of a nuclear export signal in the catalytic subunit of AMP-activated protein kinase. Mol Biol Cell. 2010;21:3433–3442. doi: 10.1091/mbc.E10-04-0347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Hawley S.A., Ross F.A., Gowans G.J., Tibarewal P., Leslie N.R., Hardie D.G. Phosphorylation by AKT within the ST loop of AMPK-α1 down-regulates its activation in tumour cells. Biochem J. 2014;459:275–287. doi: 10.1042/BJ20131344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Amodeo G.A., Rudolph M.J., Tong L. Crystal structure of the heterotrimer core of Saccharomyces cerevisiae AMPK homologue SNF1. Nature. 2007;449:492–495. doi: 10.1038/nature06127. [DOI] [PubMed] [Google Scholar]
  • 91.Townley R., Shapiro L. Crystal structures of the adenylate sensor from fission yeast AMP-activated protein kinase. Science. 2007;315:1726–1729. doi: 10.1126/science.1137503. [DOI] [PubMed] [Google Scholar]
  • 92.Xiao B., Heath R., Saiu P., Leiper F.C., Leone P., Jing C. Structural basis for AMP binding to mammalian AMP-activated protein kinase. Nature. 2007;449:496–500. doi: 10.1038/nature06161. [DOI] [PubMed] [Google Scholar]
  • 93.Koay A., Rimmer K.A., Mertens H.D.T., Gooley P.R., Stapleton D. Oligosaccharide recognition and binding to the carbohydrate binding module of AMP-activated protein kinase. FEBS Lett. 2007;581:5055–5059. doi: 10.1016/j.febslet.2007.09.044. [DOI] [PubMed] [Google Scholar]
  • 94.Hudson E.R., Pan D.A., James J., Lucocq J.M., Hawley S.A., Green K.A. A novel domain in AMP-activated protein kinase causes glycogen storage bodies similar to those seen in hereditary cardiac arrhythmias. Curr Biol. 2003;13:861–866. doi: 10.1016/s0960-9822(03)00249-5. [DOI] [PubMed] [Google Scholar]
  • 95.Polekhina G., Gupta A., Michell B.J., van Denderen B., Murthy S., Feil S.C. AMPK β subunit targets metabolic stress-sensing to glycogen. Curr Biol. 2003;13:867–871. doi: 10.1016/s0960-9822(03)00292-6. [DOI] [PubMed] [Google Scholar]
  • 96.Koay A., Woodcroft B., Petrie E.J., Yue H., Emanuelle S., Bieri M. AMPK β subunits display isoform specific affinities for carbohydrates. FEBS Lett. 2010;584:3499–3503. doi: 10.1016/j.febslet.2010.07.015. [DOI] [PubMed] [Google Scholar]
  • 97.Polekhina G., Gupta A., van Denderen B.J.W., Feil S.C., Kemp B.E., Stapleton D. Structural basis for glycogen recognition by AMP-activated protein kinase. Structure. 2005;13:1453–1462. doi: 10.1016/j.str.2005.07.008. [DOI] [PubMed] [Google Scholar]
  • 98.Mobbs J.I., Koay A., di Paolo A., Bieri M., Petrie E.J., Gorman M.A. Determinants of oligosaccharide specificity of the carbohydrate binding modules of AMP-activated protein kinase. Biochem J. 2015;468:245–257. doi: 10.1042/BJ20150270. [DOI] [PubMed] [Google Scholar]
  • 99.Oligschlaeger Y., Miglianico M., Chanda D., Scholz R., Thali R.F., Tuerk R. The recruitment of AMP-activated protein kinase to glycogen is regulated by autophosphorylation. J Biol Chem. 2015;290:11715–11728. doi: 10.1074/jbc.M114.633271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Emanuelle S., Hossain M.I., Moller I.E., Pedersen H.L., van de Meene A.M., Doblin M.S. SnRK1 from Arabidopsis thaliana is an atypical AMPK. Plant J. 2015;82:183–192. doi: 10.1111/tpj.12813. [DOI] [PubMed] [Google Scholar]
  • 101.Carling D., Hardie D.G. The substrate and sequence specificity of the AMP-activated protein kinase. Phosphorylation of glycogen synthase and phosphorylase kinase. Biochim Biophys Acta. 1989;1012:81–86. doi: 10.1016/0167-4889(89)90014-1. [DOI] [PubMed] [Google Scholar]
  • 102.Jørgensen S.B., Nielsen J.N., Birk J.B., Olsen G.S., Viollet B., Andreelli F. The α2-5′AMP-activated protein kinase is a site 2 glycogen synthase kinase in skeletal muscle and is responsive to glucose loading. Diabetes. 2004;53:3074–3081. doi: 10.2337/diabetes.53.12.3074. [DOI] [PubMed] [Google Scholar]
  • 103.Bultot L., Guigas B., von Wilamowitz-Moellendorff A., Maisin L., Vertommen D., Hussain N. AMP-activated protein kinase phosphorylates and inactivates liver glycogen synthase. Biochem J. 2012;443:193–203. doi: 10.1042/BJ20112026. [DOI] [PubMed] [Google Scholar]
  • 104.McBride A., Hardie D.G. AMP-activated protein kinase-a sensor of glycogen as well as AMP and ATP? Acta Physiol. 2009;196:99–113. doi: 10.1111/j.1748-1716.2009.01975.x. [DOI] [PubMed] [Google Scholar]
  • 105.McBride A., Ghilagaber S., Nikolaev A., Hardie D.G. The glycogen-binding domain on AMPK β subunit allows the kinase to act as a glycogen sensor. Cell Metab. 2009;9:23–34. doi: 10.1016/j.cmet.2008.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Bateman A. The structure of a domain common to archaebacteria and the homocystinuria disease protein. Trends Biochem Sci. 1997;22:12–13. doi: 10.1016/s0968-0004(96)30046-7. [DOI] [PubMed] [Google Scholar]
  • 107.Scott J.W., Hawley S.A., Green K.A., Anis M., Stewart G., Scullion G.A. CBS domains form energy-sensing modules whose binding of adenosine ligands is disrupted by disease mutations. J Clin Invest. 2004;113:274–284. doi: 10.1172/JCI19874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Chen L., Wang J., Zhang Y.Y., Yan S.F., Neumann D., Schlattner U. AMP-activated protein kinase undergoes nucleotide-dependent conformational changes. Nat Struct Mol Biol. 2012;19:716–718. doi: 10.1038/nsmb.2319. [DOI] [PubMed] [Google Scholar]
  • 109.Xin F.J., Wang J., Zhao R.Q., Wang Z.X., Wu J.W. Coordinated regulation of AMPK activity by multiple elements in the α-subunit. Cell Res. 2013;23:1237–1240. doi: 10.1038/cr.2013.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Riek U., Scholz R., Konarev P., Rufer A., Suter M., Nazabal A. Structural properties of AMP-activated protein kinase: dimerization, molecular shape, and changes upon ligand binding. J Biol Chem. 2008;283:18331–18343. doi: 10.1074/jbc.M708379200. [DOI] [PubMed] [Google Scholar]
  • 111.Oakhill J.S., Chen Z.P., Scott J.W., Steel R., Castelli L.A., Ling N.M. β-Subunit myristoylation is the gatekeeper for initiating metabolic stress sensing by AMP-activated protein kinase (AMPK) Proc Natl Acad Sci U S A. 2010;107:19237–19241. doi: 10.1073/pnas.1009705107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Leech A., Nath N., McCartney R.R., Schmidt M.C. Isolation of mutations in the catalytic domain of the SNF1 kinase that render its activity independent of the SNF4 subunit. Eukaryot Cell. 2003;2:265–273. doi: 10.1128/EC.2.2.265-273.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Chandrashekarappa D.G., McCartney R.R., Schmidt M.C. Ligand binding to the AMP-activated protein kinase active site mediates protection of the activation loop from dephosphorylation. J Biol Chem. 2013;288:89–98. doi: 10.1074/jbc.M112.422659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Zhang C.S., Jiang B., Li M.Q., Zhu M.J., Peng Y.Y., Zhang Y.L. The lysosomal v-ATPase-regulator complex is a common activator for AMPK and mTORC1, acting as a switch between catabolism and anabolism. Cell Metab. 2014;20:526–540. doi: 10.1016/j.cmet.2014.06.014. [DOI] [PubMed] [Google Scholar]
  • 115.Zhang Y.L., Guo H.L., Zhang C.S., Lin S.Y., Yin Z.Y., Peng Y.Y. AMP as a low-energy charge signal autonomously initiates assembly of AXIN-AMPK-LKB1 complex for AMPK activation. Cell Metab. 2013;18:546–555. doi: 10.1016/j.cmet.2013.09.005. [DOI] [PubMed] [Google Scholar]
  • 116.Horman S., Vertommen D., Heath R., Neumann D., Mouton V., Woods A. Insulin antagonizes ischemia-induced Thr172 phosphorylation of AMP-activated protein kinase α-subunits in heart via hierarchical phosphorylation of Ser485/491. J Biol Chem. 2006;281:5335–5340. doi: 10.1074/jbc.M506850200. [DOI] [PubMed] [Google Scholar]
  • 117.Hurley R.L., Barré L.K., Wood S.D., Anderson K.A., Kemp B.E., Means A.R. Regulation of AMP-activated protein kinase by multisite phosphorylation in response to agents that elevate cellular cAMP. J Biol Chem. 2006;281:36662–36672. doi: 10.1074/jbc.M606676200. [DOI] [PubMed] [Google Scholar]
  • 118.Djouder N., Tuerk R.D., Suter M., Salvioni P., Thali R.F., Scholz R. PKA phosphorylates and inactivates AMPKα to promote efficient lipolysis. EMBO J. 2010;29:469–481. doi: 10.1038/emboj.2009.339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Suzuki T., Bridges D., Nakada D., Skiniotis G., Morrison S.J., Lin J.D. Inhibition of AMPK catabolic action by GSK3. Mol Cell. 2013;50:407–419. doi: 10.1016/j.molcel.2013.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Serrill J.D., Tan M., Fotso S., Sikorska J., Kasanah N., Hau A.M. Apoptolidins A and C activate AMPK in metabolically sensitive cell types and are mechanistically distinct from oligomycin A. Biochem Pharmacol. 2015;93:251–265. doi: 10.1016/j.bcp.2014.11.015. [DOI] [PubMed] [Google Scholar]
  • 121.Tang X., Zhuang J.J., Chen J., Yu L., Hu L.H., Jiang H.L. Arctigenin efficiently enhanced sedentary mice treadmill endurance. PLoS One. 2011;6:e24224. doi: 10.1371/journal.pone.0024224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Lee Y.S., Kim W.S., Kim K.H., Yoon M.J., Cho H.J., Shen Y. Berberine, a natural plant product, activates AMP-activated protein kinase with beneficial metabolic effects in diabetic and insulin-resistant states. Diabetes. 2006;55:2256–2264. doi: 10.2337/db06-0006. [DOI] [PubMed] [Google Scholar]
  • 123.Wu W.D., Hu Z.M., Shang M.J., Zhao D.J., Zhang C.W., Hong D.F. Cordycepin down-regulates multiple drug resistant (MDR)/HIF-1α through regulating AMPK/mTORC1 signaling in GBC-SD gallbladder cancer cells. Int J Mol Sci. 2014;15:12778–12790. doi: 10.3390/ijms150712778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Xiao N., Mei F., Sun Y., Pan G.J., Liu B.L., Liu K. Quercetin, luteolin, and epigallocatechin gallate promote glucose disposal in adipocytes with regulation of AMP-activated kinase and/or sirtuin 1 activity. Planta Med. 2014;80:993–1000. doi: 10.1055/s-0034-1382864. [DOI] [PubMed] [Google Scholar]
  • 125.Baur J.A., Pearson K.J., Price N.L., Jamieson H.A., Lerin C., Kalra A. Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006;444:337–342. doi: 10.1038/nature05354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Shrotriya S., Tyagi A., Deep G., Orlicky D.J., Wisell J., Wang X.J. Grape seed extract and resveratrol prevent 4-nitroquinoline 1-oxide induced oral tumorigenesis in mice by modulating AMPK activation and associated biological responses. Mol Carcinog. 2015;54:291–300. doi: 10.1002/mc.22099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Yeung E.D., Morrison A., Plumeri D., Wang J.Y., Tong C., Yan X.Y. Alternol exerts prostate-selective antitumor effects through modulations of the AMPK signaling pathway. Prostate. 2012;72:165–172. doi: 10.1002/pros.21417. [DOI] [PubMed] [Google Scholar]
  • 128.Hwang Y.P., Choi J.H., Han E.H., Kim H.G., Wee J.H., Jung K.O. Purple sweet potato anthocyanins attenuate hepatic lipid accumulation through activating adenosine monophosphate-activated protein kinase in human HepG2 cells and obese mice. Nutr Res. 2011;31:896–906. doi: 10.1016/j.nutres.2011.09.026. [DOI] [PubMed] [Google Scholar]
  • 129.Ullah I., Park H.Y., Kim M.O. Anthocyanins protect against kainic acid-induced excitotoxicity and apoptosis via ROS-activated AMPK pathway in hippocampal neurons. CNS Neurosci Ther. 2014;20:327–338. doi: 10.1111/cns.12218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Ono M., Fujimori K. Antiadipogenic effect of dietary apigenin through activation of AMPK in 3T3-L1 cells. J Agric Food Chem. 2011;59:13346–13352. doi: 10.1021/jf203490a. [DOI] [PubMed] [Google Scholar]
  • 131.Tan W.Q., Chen G., Jia B., Ye M. Artemisinin inhibits neuroblastoma proliferation through activation of AHP-activated protein kinase (AMPK) signaling. Pharmazie. 2014;69:468–472. [PubMed] [Google Scholar]
  • 132.Son M.J., Minakawa M., Miura Y., Yagasaki K. Aspalathin improves hyperglycemia and glucose intolerance in obese diabetic ob/ob mice. Eur J Nutr. 2013;52:1607–1619. doi: 10.1007/s00394-012-0466-6. [DOI] [PubMed] [Google Scholar]
  • 133.Dang Y., Ling S., Duan J., Ma J., Ni R., Xu J.W. Bavachalcone-induced manganese superoxide dismutase expression through the AMP-activated protein kinase pathway in human endothelial cells. Pharmacology. 2015;95:105–110. doi: 10.1159/000375452. [DOI] [PubMed] [Google Scholar]
  • 134.Liao C.C., Ou T.T., Huang H.P., Wang C.J. The inhibition of oleic acid induced hepatic lipogenesis and the promotion of lipolysis by caffeic acid via up-regulation of AMP-activated kinase. J Sci Food Agric. 2014;94:1154–1162. doi: 10.1002/jsfa.6386. [DOI] [PubMed] [Google Scholar]
  • 135.Chiang E.P.I., Tsai S.Y., Kuo Y.H., Pai M.H., Chiu H.L., Rodriguez R.L. Caffeic acid derivatives inhibit the growth of colon cancer: involvement of the PI3-K/AKT and AMPK signaling pathways. PLoS One. 2014;9:e99631. doi: 10.1371/journal.pone.0099631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Kim J.H., Lee J.O., Lee S.K., Kim N., You G.Y., Moon J.W. Celastrol suppresses breast cancer MCF-7 cell viability via the AMP-activated protein kinase (AMPK)-induced p53-polo like kinase 2 (PLK-2) pathway. Cell Signal. 2013;25:805–813. doi: 10.1016/j.cellsig.2012.12.005. [DOI] [PubMed] [Google Scholar]
  • 137.Zhang T.S., Yamamoto N., Ashida H. Chalcones suppress fatty acid-induced lipid accumulation through a LKB1/AMPK signaling pathway in HepG2 cells. Food Funct. 2014;5:1134–1141. doi: 10.1039/c3fo60694e. [DOI] [PubMed] [Google Scholar]
  • 138.Chiu C.Y., Chan I.L., Yang T.H., Liu S.H., Chiang M.T. Supplementation of chitosan alleviates high-fat diet-enhanced lipogenesis in rats via adenosine monophosphate (AMP)-activated protein kinase activation and inhibition of lipogenesis-associated genes. J Agric Food Chem. 2015;63:2979–2988. doi: 10.1021/acs.jafc.5b00198. [DOI] [PubMed] [Google Scholar]
  • 139.Shao J.J., Zhang A.P., Qin W., Zheng L., Zhu Y.F., Chen X. AMP-activated protein kinase (AMPK) activation is involved in chrysin-induced growth inhibition and apoptosis in cultured A549 lung cancer cells. Biochem Biophys Res Commun. 2012;423:448–453. doi: 10.1016/j.bbrc.2012.05.123. [DOI] [PubMed] [Google Scholar]
  • 140.Chen X.B., Zhuang J.J., Liu J.H., Lei M., Ma L., Chen J. Potential AMPK activators of cucurbitane triterpenoids from Siraitia grosvenorii Swingle. Bioorg Med Chem. 2011;19:5776–5781. doi: 10.1016/j.bmc.2011.08.030. [DOI] [PubMed] [Google Scholar]
  • 141.Kubota M., Shimizu M., Sakai H., Yasuda Y., Terakura D., Baba A. Preventive effects of curcumin on the development of azoxymethane-induced colonic preneoplastic lesions in male C57BL/KsJ-db/db obese mice. Nutr Cancer. 2012;64:72–79. doi: 10.1080/01635581.2012.630554. [DOI] [PubMed] [Google Scholar]
  • 142.Jiménez-Flores L.M., López-Briones S., Macías-Cervantes M.H., Ramírez-Emiliano J., Pérez-Vázquez V. A PPARγ, NF-κB and AMPK-dependent mechanism may be involved in the beneficial effects of curcumin in the diabetic db/db mice liver. Molecules. 2014;19:8289–8302. doi: 10.3390/molecules19068289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Park S., Kang S., Jeong D.Y., Jeong S.Y., Park J.J., Yun H.S. Cyanidin and malvidin in aqueous extracts of black carrots fermented with Aspergillus oryzae prevent the impairment of energy, lipid and glucose metabolism in estrogen-deficient rats by AMPK activation. Genes Nutr. 2015;10:455. doi: 10.1007/s12263-015-0455-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Kim S.J., Kim H.M., Lee E.S., Kim N., Lee J.O., Lee H.J. Dehydrozingerone exerts beneficial metabolic effects in high-fat diet-induced obese mice via AMPK activation in skeletal muscle. J Cell Mol Med. 2015;19:620–629. doi: 10.1111/jcmm.12455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Jin X., Chen M.L., Yi L., Chang H., Zhang T., Wang L. Delphinidin-3-glucoside protects human umbilical vein endothelial cells against oxidized low-density lipoprotein-induced injury by autophagy upregulation via the AMPK/SIRT1 signaling pathway. Mol Nutr Food Res. 2014;58:1941–1951. doi: 10.1002/mnfr.201400161. [DOI] [PubMed] [Google Scholar]
  • 146.Mandal S., Mukhopadhyay S., Bandhopadhyay S., Sen G., Biswas T. 14-Deoxyandrographolide alleviates ethanol-induced hepatosteatosis through stimulation of AMP-activated protein kinase activity in rats. Alcohol. 2014;48:123–132. doi: 10.1016/j.alcohol.2013.11.005. [DOI] [PubMed] [Google Scholar]
  • 147.Zhao Z.Q., Yin J.Q., Wu M.S., Song G.H., Xie X.B., Zou C.Y. Dihydromyricetin activates AMP-activated protein kinase and P38MAPK exerting antitumor potential in osteosarcoma. Cancer Prev Res. 2014;7:927–938. doi: 10.1158/1940-6207.CAPR-14-0067. [DOI] [PubMed] [Google Scholar]
  • 148.Ladurner A., Atanasov A.G., Heiss E.H., Baumgartner L., Schwaiger S., Rollinger J.M. 2-(2,4-Dihydroxyphenyl)-5-(E)-propenylbenzofuran promotes endothelial nitric oxide synthase activity in human endothelial cells. Biochem Pharmacol. 2012;84:804–812. doi: 10.1016/j.bcp.2012.06.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Chen Z.F., Zhang L., Yi J.Y., Yang Z.B., Zhang Z.J., Li Z. Promotion of adiponectin multimerization by emodin: a novel AMPK activator with PPARγ-agonist activity. J Cell Biochem. 2012;113:3547–3558. doi: 10.1002/jcb.24232. [DOI] [PubMed] [Google Scholar]
  • 150.Tzeng T.F., Lu H.J., Liou S.S., Chang C.J., Liu I.M. Emodin protects against high-fat diet-induced obesity via regulation of AMP-activated protein kinase pathways in white adipose tissue. Planta Med. 2012;78:943–950. doi: 10.1055/s-0031-1298626. [DOI] [PubMed] [Google Scholar]
  • 151.Tzeng T.F., Lu H.J., Liou S.S., Chang C.J., Liu I.M. Emodin, a naturally occurring anthraquinone derivative, ameliorates dyslipidemia by activating AMP-activated protein kinase in high-fat-diet-fed rats. Evid Based Complement Alternat Med. 2012;2012:781812. doi: 10.1155/2012/781812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Chen C.C., Lin J.T., Cheng Y.F., Kuo C.Y., Huang C.F., Kao S.H. Amelioration of LPS-induced inflammation response in microglia by AMPK activation. Biomed Res Int. 2014;2014:692061. doi: 10.1155/2014/692061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Huang B.P., Lin C.H., Chen H.M., Lin J.T., Cheng Y.F., Kao S.H. AMPK activation inhibits expression of proinflammatory mediators through downregulation of PI3K/p38 MAPK and NF-κB signaling in murine macrophages. DNA Cell Biol. 2015;34:133–141. doi: 10.1089/dna.2014.2630. [DOI] [PubMed] [Google Scholar]
  • 154.Chen D., Pamu S., Cui Q.Z., Chan T.H., Dou Q.P. Novel epigallocatechin gallate (EGCG) analogs activate AMP-activated protein kinase pathway and target cancer stem cells. Bioorg Med Chem. 2012;20:3031–3037. doi: 10.1016/j.bmc.2012.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Kuo Y.H., Lin C.H., Shih C.C. Ergostatrien-3β-ol from Antrodia camphorata inhibits diabetes and hyperlipidemia in high-fat-diet treated mice via regulation of hepatic related genes, glucose transporter 4, and AMP-activated protein kinase phosphorylation. J Agric Food Chem. 2015;63:2479–2489. doi: 10.1021/acs.jafc.5b00073. [DOI] [PubMed] [Google Scholar]
  • 156.Jeong K.J., Kim D.Y., Quan H.Y., Jo H.K., Kim G.W., Chung S.H. Effects of eugenol on hepatic glucose production and AMPK signaling pathway in hepatocytes and C57BL/6J mice. Fitoterapia. 2014;93:150–162. doi: 10.1016/j.fitote.2013.12.023. [DOI] [PubMed] [Google Scholar]
  • 157.Lee Y.S., Cha B.Y., Choi S.S., Harada Y., Choi B.K., Yonezawa T. Fargesin improves lipid and glucose metabolism in 3T3-L1 adipocytes and high-fat diet-induced obese mice. Biofactors. 2012;38:300–308. doi: 10.1002/biof.1022. [DOI] [PubMed] [Google Scholar]
  • 158.Seo J.B., Park S.W., Choe S.S., Jeong H.W., Park J.Y., Choi E.W. Foenumoside B from Lysimachia foenum-graecum inhibits adipocyte differentiation and obesity induced by high-fat diet. Biochem Biophys Res Commun. 2012;417:800–806. doi: 10.1016/j.bbrc.2011.12.039. [DOI] [PubMed] [Google Scholar]
  • 159.Kawaguchi T., Hayakawa M., Koga H., Torimura T. Effects of fucoidan on proliferation, AMP-activated protein kinase, and downstream metabolism- and cell cycle-associated molecules in poorly differentiated human hepatoma HLF cells. Int J Oncol. 2015;46:2216–2222. doi: 10.3892/ijo.2015.2928. [DOI] [PubMed] [Google Scholar]
  • 160.Chen M.H., Lin C.H., Shih C.C. Antidiabetic and antihyperlipidemic effects of Clitocybe nuda on glucose transporter 4 and AMP-activated protein kinase phosphorylation in high-fat-fed mice. Evid Based Complement Alternat Med. 2014;2014:981046. doi: 10.1155/2014/981046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Shih C.C., Chen M.H., Lin C.H. Validation of the antidiabetic and hypolipidemic effects of Clitocybe nuda by assessment of glucose transporter 4 and gluconeogenesis and AMPK phosphorylation in streptozotocin-induced mice. Evid Based Complement Alternat Med. 2014;2014:705636. doi: 10.1155/2014/705636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Doan K.V., Ko C.M., Kinyua A.W., Yang D.J., Choi Y.H., Oh I.Y. Gallic acid regulates body weight and glucose homeostasis through AMPK activation. Endocrinology. 2015;156:157–168. doi: 10.1210/en.2014-1354. [DOI] [PubMed] [Google Scholar]
  • 163.Kim S.H., Park E.J., Lee C.R., Chun J.N., Cho N.H., Kim I.G. Geraniol induces cooperative interaction of apoptosis and autophagy to elicit cell death in PC-3 prostate cancer cells. Int J Oncol. 2012;40:1683–1690. doi: 10.3892/ijo.2011.1318. [DOI] [PubMed] [Google Scholar]
  • 164.Shin S.S., Park D., Lee H.Y., Hong Y., Choi J., Oh J. The herbal composition GGEx18 from Laminaria japonica, Rheum palmatum, and Ephedra sinica reduces obesity via skeletal muscle AMPK and PPARα. Pharm Biol. 2012;50:506–515. doi: 10.3109/13880209.2011.618502. [DOI] [PubMed] [Google Scholar]
  • 165.Fan J.Z., Yang X., Bi Z.G. 6-Gingerol inhibits osteosarcoma cell proliferation through apoptosis and AMPK activation. Tumour Biol. 2015;36:1135–1141. doi: 10.1007/s13277-014-2723-1. [DOI] [PubMed] [Google Scholar]
  • 166.Lee J.O., Kim N., Lee H.J., Moon J.W., Lee S.K., Kim S.J. [6]-Gingerol affects glucose metabolism by dual regulation via the AMPKα2-mediated AS160-Rab5 pathway and AMPK-mediated insulin sensitizing effects. J Cell Biochem. 2015;116:1401–1410. doi: 10.1002/jcb.25100. [DOI] [PubMed] [Google Scholar]
  • 167.Li Y., Tran V.H., Koolaji N., Duke C., Roufogalis B.D. (S)-[6]-Gingerol enhances glucose uptake in L6 myotubes by activation of AMPK in response to [Ca2+]i. J Pharm Pharm Sci. 2013;16:304–312. doi: 10.18433/j34g7p. [DOI] [PubMed] [Google Scholar]
  • 168.Yuan H.D., Kim do Y., Quan H.Y., Kim S.J., Jung M.S., Chung S.H. Ginsenoside Rg2 induces orphan nuclear receptor SHP gene expression and inactivates GSK3β via AMP-activated protein kinase to inhibit hepatic glucose production in HepG2 cells. Chem Biol Interact. 2012;195:35–42. doi: 10.1016/j.cbi.2011.10.006. [DOI] [PubMed] [Google Scholar]
  • 169.Kim D.Y., Park Y.G., Quan H.Y., Kim S.J., Jung M.S., Chung S.H. Ginsenoside Rd stimulates the differentiation and mineralization of osteoblastic MC3T3-E1 cells by activating AMP-activated protein kinase via the BMP-2 signaling pathway. Fitoterapia. 2012;83:215–222. doi: 10.1016/j.fitote.2011.10.017. [DOI] [PubMed] [Google Scholar]
  • 170.Quan H.Y., Yuan H.D., Jung M.S., Ko S.K., Park Y.G., Chung S.H. Ginsenoside Re lowers blood glucose and lipid levels via activation of AMP-activated protein kinase in HepG2 cells and high-fat diet fed mice. Int J Mol Med. 2011;29:73–80. doi: 10.3892/ijmm.2011.805. [DOI] [PubMed] [Google Scholar]
  • 171.Kim M.J., Yun H., Kim D.H., Kang I., Choe W., Kim S.S. AMP-activated protein kinase determines apoptotic sensitivity of cancer cells to ginsenoside-Rh2. J Ginseng Res. 2014;38:16–21. doi: 10.1016/j.jgr.2013.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Shin D.J., Kim J.E., Lim T.G., Jeong E.H., Park G., Kang N.J. 20-O-β-D-glucopyranosyl-20(S)-protopanaxadiol suppresses UV-induced MMP-1 expression through AMPK-mediated mTOR inhibition as a downstream of the PKA-LKB1 pathway. J Cell Biochem. 2014;115:1702–1711. doi: 10.1002/jcb.24833. [DOI] [PubMed] [Google Scholar]
  • 173.Lee J.W., Choe S.S., Jang H., Kim J., Jeong H.W., Jo H. AMPK activation with glabridin ameliorates adiposity and lipid dysregulation in obesity. J Lipid Res. 2012;53:1277–1286. doi: 10.1194/jlr.M022897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Sawada K., Yamashita Y., Zhang T.S., Nakagawa K., Ashida H. Glabridin induces glucose uptake via the AMP-activated protein kinase pathway in muscle cells. Mol Cell Endocrinol. 2014;393:99–108. doi: 10.1016/j.mce.2014.06.009. [DOI] [PubMed] [Google Scholar]
  • 175.Banerjee S., Ghoshal S., Porter T.D. Phosphorylation of hepatic AMP-activated protein kinase and liver kinase B1 is increased after a single oral dose of green tea extract to mice. Nutr Res. 2012;32:985–990. doi: 10.1016/j.nutres.2012.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Huang H.C., Lin J.K. Pu-erh tea, green tea, and black tea suppresses hyperlipidemia, hyperleptinemia and fatty acid synthase through activating AMPK in rats fed a high-fructose diet. Food Funct. 2012;3:170–177. doi: 10.1039/c1fo10157a. [DOI] [PubMed] [Google Scholar]
  • 177.Lin Y.C., Hung C.M., Tsai J.C., Lee J.C., Chen Y.L.S., Wei C.W. Hispidulin potently inhibits human glioblastoma multiforme cells through activation of AMP-activated protein kinase (AMPK) J Agric Food Chem. 2010;58:9511–9517. doi: 10.1021/jf1019533. [DOI] [PubMed] [Google Scholar]
  • 178.Yang J.M., Hung C.M., Fu C.N., Lee J.C., Huang C.H., Yang M.H. Hispidulin sensitizes human ovarian cancer cells to TRAIL-induced apoptosis by AMPK activation leading to Mcl-1 block in translation. J Agric Food Chem. 2010;58:10020–10026. doi: 10.1021/jf102304g. [DOI] [PubMed] [Google Scholar]
  • 179.Wang Y.G., Liu W.P., He X.S., Fei Z. Hispidulin enhances the anti-tumor effects of temozolomide in glioblastoma by activating AMPK. Cell Biochem Biophys. 2015;71:701–706. doi: 10.1007/s12013-014-0252-6. [DOI] [PubMed] [Google Scholar]
  • 180.Kapoor S. Attenuation of tumor growth by honokiol: an evolving role in oncology. Drug Discov Ther. 2012;6:327–328. doi: 10.5582/ddt.2012.v6.6.327. [DOI] [PubMed] [Google Scholar]
  • 181.Seo M.S., Kim J.H., Kim H.J., Chang K.C., Park S.W. Honokiol activates the LKB1-AMPK signaling pathway and attenuates the lipid accumulation in hepatocytes. Toxicol Appl Pharmacol. 2015;284:113–124. doi: 10.1016/j.taap.2015.02.020. [DOI] [PubMed] [Google Scholar]
  • 182.Yin J.J., Luo Y.Q., Deng H.L., Qin S.M., Tang W.J., Zeng L. Hugan Qingzhi medication ameliorates hepatic steatosis by activating AMPK and PPARα pathways in L02 cells and HepG2 cells. J Ethnopharmacol. 2014;154:229–239. doi: 10.1016/j.jep.2014.04.011. [DOI] [PubMed] [Google Scholar]
  • 183.Yuan T., Nahar P., Sharma M., Liu K., Slitt A., Aisa H.A. Indazole-type alkaloids from Nigella sativa seeds exhibit antihyperglycemic effects via AMPK activation in vitro. J Nat Prod. 2014;77:2316–2320. doi: 10.1021/np500398m. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Zhou J.X., Yoshitomi H., Liu T.H., Zhou B.X., Sun W., Qin L.L. Isoquercitrin activates the AMP-activated protein kinase (AMPK) signal pathway in rat H4IIE cells. BMC Complement Altern Med. 2014;14:42. doi: 10.1186/1472-6882-14-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Dong G.Z., Lee J.H., Ki S.H., Yang J.H., Cho I.J., Kang S.H. AMPK activation by isorhamnetin protects hepatocytes against oxidative stress and mitochondrial dysfunction. Eur J Pharmacol. 2014;740:634–640. doi: 10.1016/j.ejphar.2014.06.017. [DOI] [PubMed] [Google Scholar]
  • 186.Wang D.K., Tian M., Qi Y., Chen G., Xu L.J., Zou X. Jinlida granule inhibits palmitic acid induced-intracellular lipid accumulation and enhances autophagy in NIT-1 pancreatic β cells through AMPK activation. J Ethnopharmacol. 2015;161:99–107. doi: 10.1016/j.jep.2014.12.005. [DOI] [PubMed] [Google Scholar]
  • 187.Qian Q., Liu X.F., He W., An Y.T., Chen Q., Wu J.X. TG accumulation inhibitory effects of Jinqi formula by AMPK signaling pathway. J Ethnopharmacol. 2012;143:41–48. doi: 10.1016/j.jep.2012.05.052. [DOI] [PubMed] [Google Scholar]
  • 188.Jaiswal N., Yadav P.P., Maurya R., Srivastava A.K., Tamrakar A.K. Karanjin from Pongamia pinnata induces GLUT4 translocation in skeletal muscle cells in a phosphatidylinositol-3-kinase-independent manner. Eur J Pharmacol. 2011;670:22–28. doi: 10.1016/j.ejphar.2011.08.049. [DOI] [PubMed] [Google Scholar]
  • 189.Kim H.S., Lim J., Lee da Y., Ryu J.H., Lim J.S. Kazinol C from Broussonetia kazinoki activates AMP-activated protein kinase to induce antitumorigenic effects in HT-29 colon cancer cells. Oncol Rep. 2015;33:223–229. doi: 10.3892/or.2014.3601. [DOI] [PubMed] [Google Scholar]
  • 190.Han J.Y., Park S.H., Yang J.H., Kim M.G., Cho S.S., Yoon G. Licochalcone suppresses LXRα-induced hepatic lipogenic gene expression through AMPK/Sirt1 pathway activation. Toxicol Res. 2014;30:19–25. doi: 10.5487/tr.2014.30.1.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Jeong G.S., Lee D.S., Li B., Kim J.J., Kim E.C., Kim Y.C. Anti-inflammatory effects of lindenenyl acetate via heme oxygenase-1 and AMPK in human periodontal ligament cells. Eur J Pharmacol. 2011;670:295–303. doi: 10.1016/j.ejphar.2011.08.008. [DOI] [PubMed] [Google Scholar]
  • 192.Niu Y.C., Li S.T., Na L.X., Feng R.N., Liu L.Y., Li Y. Mangiferin decreases plasma free fatty acids through promoting its catabolism in liver by activation of AMPK. PLoS One. 2012;7:e30782. doi: 10.1371/journal.pone.0030782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Wan Y., Liu L.Y., Hong Z.F., Peng J. Ethanol extract of Cirsium japonicum attenuates hepatic lipid accumulation via AMPK activation in human HepG2 cells. Exp Ther Med. 2014;8:79–84. doi: 10.3892/etm.2014.1698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Han J., Yi J., Liang F.Y., Jiang B., Xiao Y., Gao S.H. X-3, a mangiferin derivative, stimulates AMP-activated protein kinase and reduces hyperglycemia and obesity in db/db mice. Mol Cell Endocrinol. 2015;405:63–73. doi: 10.1016/j.mce.2015.02.008. [DOI] [PubMed] [Google Scholar]
  • 195.Song J.N., Li J., Hou F.J., Wang X.N., Liu B.L. Mangiferin inhibits endoplasmic reticulum stress-associated thioredoxin-interacting protein/NLRP3 inflammasome activation with regulation of AMPK in endothelial cells. Metabolism. 2015;64:428–437. doi: 10.1016/j.metabol.2014.11.008. [DOI] [PubMed] [Google Scholar]
  • 196.Chen Y.Y., Lee M.H., Hsu C.C., Wei C.L., Tsai Y.C. Methyl cinnamate inhibits adipocyte differentiation via activation of the CaMKK2-AMPK pathway in 3T3-L1 preadipocytes. J Agric Food Chem. 2012;60:955–963. doi: 10.1021/jf203981x. [DOI] [PubMed] [Google Scholar]
  • 197.Kim S., Ka S.O., Lee Y., Park B.H., Fei X., Jung J.K. The new 4-O-methylhonokiol analog GS12021 inhibits inflammation and macrophage chemotaxis: role of AMP-activated protein kinase α activation. PLoS One. 2015;10:e0117120. doi: 10.1371/journal.pone.0117120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Ptitsyn L.R., Nomura K., Sklyar, Ravcheeva A.B. The 1,4-naphthoquinone derivative from Pyrola rotundifolia activates AMPK phosphorylation in C2C12 myotubes. Fitoterapia. 2011;82:1285–1289. doi: 10.1016/j.fitote.2011.09.005. [DOI] [PubMed] [Google Scholar]
  • 199.Hsu W.H., Chen T.H., Lee B.H., Hsu Y.W., Pan T.M. Monascin and ankaflavin act as natural AMPK activators with PPARα agonist activity to down-regulate nonalcoholic steatohepatitis in high-fat diet-fed C57BL/6 mice. Food Chem Toxicol. 2014;64:94–103. doi: 10.1016/j.fct.2013.11.015. [DOI] [PubMed] [Google Scholar]
  • 200.Chen R.J., Hung C.M., Chen Y.L., Wu M.D., Yuan G.F., Wang Y.J. Monascuspiloin induces apoptosis and autophagic cell death in human prostate cancer cells via the AKT and AMPK signaling pathways. J Agric Food Chem. 2012;60:7185–7193. doi: 10.1021/jf3016927. [DOI] [PubMed] [Google Scholar]
  • 201.Pu P., Gao D.M., Mohamed S., Chen J., Zhang J., Zhou X.Y. Naringin ameliorates metabolic syndrome by activating AMP-activated protein kinase in mice fed a high-fat diet. Arch Biochem Biophys. 2012;518:61–70. doi: 10.1016/j.abb.2011.11.026. [DOI] [PubMed] [Google Scholar]
  • 202.Hien T.T., Ki S.H., Yang J.W., Oh W.K., Kang K.W. Nectandrin B suppresses the expression of adhesion molecules in endothelial cells: role of AMP-activated protein kinase activation. Food Chem Toxicol. 2014;66:286–294. doi: 10.1016/j.fct.2014.01.052. [DOI] [PubMed] [Google Scholar]
  • 203.Hien T.T., Oh W.K., Nguyen P.H., Oh S.J., Lee M.Y., Kang K.W. Nectandrin B activates endothelial nitric-oxide synthase phosphorylation in endothelial cells: role of the AMP-activated protein kinase/estrogen receptor α/phosphatidylinositol 3-kinase/AKT pathway. Mol Pharmacol. 2011;80:1166–1178. doi: 10.1124/mol.111.073502. [DOI] [PubMed] [Google Scholar]
  • 204.Ki S.H., Lee J.W., Lim S.C., Hien T.T., Im J.H., Oh W.K. Protective effect of nectandrin B, a potent AMPK activator on neointima formation: inhibition of Pin1 expression through AMPK activation. Br J Pharmacol. 2013;168:932–945. doi: 10.1111/j.1476-5381.2012.02228.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Lee S.H., Kang S.M., Ko S.C., Lee D.H., Jeon Y.J. Octaphlorethol A, a novel phenolic compound isolated from a brown alga, Ishige foliacea, increases glucose transporter 4-mediated glucose uptake in skeletal muscle cells. Biochem Biophys Res Commun. 2012;420:576–581. doi: 10.1016/j.bbrc.2012.03.036. [DOI] [PubMed] [Google Scholar]
  • 206.Liu J., Zheng L.H., Wu N., Ma L.N., Zhong J.T., Liu G. Oleanolic acid induces metabolic adaptation in cancer cells by activating the AMP-activated protein kinase pathway. J Agric Food Chem. 2014;62:5528–5537. doi: 10.1021/jf500622p. [DOI] [PubMed] [Google Scholar]
  • 207.Lee W.H., Lin R.J., Lin S.Y., Chen Y.C., Lin H.M., Liang Y.C. Osthole enhances glucose uptake through activation of AMP-activated protein kinase in skeletal muscle cells. J Agric Food Chem. 2011;59:12874–12881. doi: 10.1021/jf2036559. [DOI] [PubMed] [Google Scholar]
  • 208.Lee W.H., Wu H.H., Huang W.J., Li Y.N., Lin R.J., Lin S.Y. N-hydroxycinnamide derivatives of osthole ameliorate hyperglycemia through activation of AMPK and p38 MAPK. Molecules. 2015;20:4516–4529. doi: 10.3390/molecules20034516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Lu C., Wang W.W., Jia Y.S., Liu X.D., Tong Z.S., Li B.H. Inhibition of AMPK/autophagy potentiates parthenolide-induced apoptosis in human breast cancer cells. J Cell Biochem. 2014;115:1458–1466. doi: 10.1002/jcb.24808. [DOI] [PubMed] [Google Scholar]
  • 210.Zou B., Ge Z.Z., Zhang Y., Du J., Xu Z., Li C.M. Persimmon tannin accounts for hypolipidemic effects of persimmon through activating of AMPK and suppressing NF-κB activation and inflammatory responses in high-fat diet rats. Food Funct. 2014;5:1536–1546. doi: 10.1039/c3fo60635j. [DOI] [PubMed] [Google Scholar]
  • 211.Adachi Y., Kanbayashi Y., Harata I., Ubagai R., Takimoto T., Suzuki K. Petasin activates AMP-activated protein kinase and modulates glucose metabolism. J Nat Prod. 2014;77:1262–1269. doi: 10.1021/np400867m. [DOI] [PubMed] [Google Scholar]
  • 212.Ryu J., Kim M.J., Kim T.O., Huh T.L., Lee S.E. Piperlongumine as a potential activator of AMP-activated protein kinase in HepG2 cells. Nat Prod Res. 2014;28:2040–2043. doi: 10.1080/14786419.2014.919283. [DOI] [PubMed] [Google Scholar]
  • 213.Kim D.Y., Kim M.S., Sa B.K., Kim M.B., Hwang J.K. Boesenbergia pandurata attenuates diet-induced obesity by activating AMP-activated protein kinase and regulating lipid metabolism. Int J Mol Sci. 2012;13:994–1005. doi: 10.3390/ijms13010994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Kang H., Koppula S. Houttuynia cordata attenuates lipid accumulation via activation of AMP-activated protein kinase signaling pathway in HepG2 cells. Am J Chin Med. 2014;42:651–664. doi: 10.1142/S0192415X14500426. [DOI] [PubMed] [Google Scholar]
  • 215.Shin J.A., Kwon K.H., Cho S.D. AMPK-activated protein kinase activation by Impatiens balsamina L. is related to apoptosis in HSC-2 human oral cancer cells. Pharmacogn Mag. 2015;11:136–142. doi: 10.4103/0973-1296.149728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Li W., Li Y., Wang Q., Yang Y. Crude extracts from Lycium barbarum suppress SREBP-1c expression and prevent diet-induced fatty liver through AMPK activation. Biomed Res Int. 2014;2014:196198. doi: 10.1155/2014/196198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Jeong Y.T., Song C.H. Antidiabetic activities of extract from Malva verticillata seed via the activation of AMP-activated protein kinase. J Microbiol Biotechnol. 2011;21:921–929. doi: 10.4014/jmb.1104.04015. [DOI] [PubMed] [Google Scholar]
  • 218.Kim A., Im M., Ma J.Y. Ethanol extract of Remotiflori radix induces endoplasmic reticulum stress-mediated cell death through AMPK/mTOR signaling in human prostate cancer cells. Sci Rep. 2015;5:8394. doi: 10.1038/srep08394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Lee J.O., Moon J.W., Lee S.K., Kim S.M., Kim N., Ko S.G. Rhus verniciflua extract modulates survival of MCF-7 breast cancer cells through the modulation of AMPK-pathway. Biol Pharm Bull. 2014;37:794–801. doi: 10.1248/bpb.b13-00893. [DOI] [PubMed] [Google Scholar]
  • 220.Song K.H., Lee S.H., Kim B.Y., Park A.Y., Kim J.Y. Extracts of Scutellaria baicalensis reduced body weight and blood triglyceride in db/db mice. Phytother Res. 2013;27:244–250. doi: 10.1002/ptr.4691. [DOI] [PubMed] [Google Scholar]
  • 221.Wu C.H., Ou T.T., Chang C.H., Chang X.Z., Yang M.Y., Wang C.J. The polyphenol extract from Sechium edule shoots inhibits lipogenesis and stimulates lipolysis via activation of AMPK signals in HepG2 cells. J Agric Food Chem. 2014;62:750–759. doi: 10.1021/jf404611a. [DOI] [PubMed] [Google Scholar]
  • 222.Liu Y.J., Shieh P.C., Lee J.C., Chen F.A., Lee C.H., Kuo S.C. Hypolipidemic activity of Taraxacum mongolicum associated with the activation of AMP-activated protein kinase in human HepG2 cells. Food Funct. 2014;5:1755–1762. doi: 10.1039/c4fo00183d. [DOI] [PubMed] [Google Scholar]
  • 223.Papadimitriou A., EBMI Peixoto, Silva K.C., Lopes de Faria J.M., Lopes de Faria J.B. Increase in AMPK brought about by cocoa is renoprotective in experimental diabetes mellitus by reducing NOX4/TGFβ-1 signaling. J Nutr Biochem. 2014;25:773–784. doi: 10.1016/j.jnutbio.2014.03.010. [DOI] [PubMed] [Google Scholar]
  • 224.Sung Y.Y., Kim D.S., Kim H.K. Viola mandshurica ethanolic extract prevents high-fat-diet-induced obesity in mice by activating AMP-activated protein kinase. Environ Toxicol Pharmacol. 2014;38:41–50. doi: 10.1016/j.etap.2014.04.028. [DOI] [PubMed] [Google Scholar]
  • 225.Pan C.H., Tsai C.H., Lin W.H., Chen G.Y., Wu C.H. Ethanolic extract of Vitis thunbergii exhibits lipid lowering properties via modulation of the AMPK-ACC pathway in hypercholesterolemic rabbits. Evid Based Complement Alternat Med. 2012;2012:436786. doi: 10.1155/2012/436786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Youn S.H., Lee J.S., Lee M.S., Cha E.Y., Thuong P.T., Kim J.R. Anticancer properties of pomolic acid-induced AMP-activated protein kinase activation in MCF7 human breast cancer cells. Biol Pharm Bull. 2012;35:105–110. doi: 10.1248/bpb.35.105. [DOI] [PubMed] [Google Scholar]
  • 227.Lin V.C.H., Tsai Y.C., Lin J.N., Fan L.L., Pan M.H., Ho C.T. Activation of AMPK by pterostilbene suppresses lipogenesis and cell-cycle progression in p53 positive and negative human prostate cancer cells. J Agric Food Chem. 2012;60:6399–6407. doi: 10.1021/jf301499e. [DOI] [PubMed] [Google Scholar]
  • 228.Kang O.H., Kim S.B., Mun S.H., Seo Y.S., Hwang H.C., Lee Y.M. Puerarin ameliorates hepatic steatosis by activating the PPARα and AMPK signaling pathways in hepatocytes. Int J Mol Med. 2015;35:803–809. doi: 10.3892/ijmm.2015.2074. [DOI] [PubMed] [Google Scholar]
  • 229.Thyagarajan-Sahu A., Lane B., Sliva D. ReishiMax, mushroom based dietary supplement, inhibits adipocyte differentiation, stimulates glucose uptake and activates AMPK. BMC Complement Altern Med. 2011;11:74. doi: 10.1186/1472-6882-11-74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Khanal P., Kang B.S., Yun H.J., Cho H.G., Makarieva T.N., Choi H.S. Aglycon of rhizochalin from the Rhizochalina incrustata induces apoptosis via activation of AMP-activated protein kinase in HT-29 colon cancer cells. Biol Pharm Bull. 2011;34:1553–1558. doi: 10.1248/bpb.34.1553. [DOI] [PubMed] [Google Scholar]
  • 231.Lee N.Y., Park K.Y., Min H.J., Song K.Y., Lim Y.Y., Park J. Inhibitory effect of vitamin U (S-methylmethionine sulfonium chloride) on differentiation in 3T3-L1 pre-adipocyte cell lines. Ann Dermatol. 2012;24:39–44. doi: 10.5021/ad.2012.24.1.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Liu Z.B., Li X.S., Simoneau A.R., Jafari M., Zi X.L. Rhodiola rosea extracts and salidroside decrease the growth of bladder cancer cell lines via inhibition of the mTOR pathway and induction of autophagy. Mol Carcinog. 2012;51:257–267. doi: 10.1002/mc.20780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Ge Y.Q., Xu X.F., Yang B., Chen Z., Cheng R.B. Saponins from Rubus parvifolius L. induce apoptosis in human chronic myeloid leukemia cells through AMPK activation and STAT3 inhibition. Asian Pac J Cancer Prev. 2014;15:5455–5461. doi: 10.7314/apjcp.2014.15.13.5455. [DOI] [PubMed] [Google Scholar]
  • 234.Lee H.I., Yun K.W., Seo K.I., Kim M.J., Lee M.K. Scopoletin prevents alcohol-induced hepatic lipid accumulation by modulating the AMPK-SREBP pathway in diet-induced obese mice. Metabolism. 2014;63:593–601. doi: 10.1016/j.metabol.2014.01.003. [DOI] [PubMed] [Google Scholar]
  • 235.Roblet C., Doyen A., Amiot J., Pilon G., Marette A., Bazinet L. Enhancement of glucose uptake in muscular cell by soybean charged peptides isolated by electrodialysis with ultrafiltration membranes (EDUF): activation of the AMPK pathway. Food Chem. 2014;147:124–130. doi: 10.1016/j.foodchem.2013.09.108. [DOI] [PubMed] [Google Scholar]
  • 236.Choi K.M., Lee Y.S., Kim W., Kim S.J., Shin K.O., Yu J.Y. Sulforaphane attenuates obesity by inhibiting adipogenesis and activating the AMPK pathway in obese mice. J Nutr Biochem. 2014;25:201–207. doi: 10.1016/j.jnutbio.2013.10.007. [DOI] [PubMed] [Google Scholar]
  • 237.Kim M.S., Hur H.J., Kwon D.Y., Hwang J.T. Tangeretin stimulates glucose uptake via regulation of AMPK signaling pathways in C2C12 myotubes and improves glucose tolerance in high-fat diet-induced obese mice. Mol Cell Endocrinol. 2012;358:127–134. doi: 10.1016/j.mce.2012.03.013. [DOI] [PubMed] [Google Scholar]
  • 238.Hwang S.L., Yang J.H., Jeong Y.T., Kim Y.D., Li X., Lu Y. Tanshinone IIA improves endoplasmic reticulum stress-induced insulin resistance through AMP-activated protein kinase. Biochem Biophys Res Commun. 2013;430:1246–1252. doi: 10.1016/j.bbrc.2012.12.066. [DOI] [PubMed] [Google Scholar]
  • 239.Wu W.Y., Yan H., Wang X.B., Gui Y.Z., Gao F., Tang X.L. Sodium tanshinone IIA silate inhibits high glucose-induced vascular smooth muscle cell proliferation and migration through activation of AMP-activated protein kinase. PLoS One. 2014;9:e94957. doi: 10.1371/journal.pone.0094957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Park H.Y., Kunitake Y., Hirasaki N., Tanaka M., Matsui T. Theaflavins enhance intestinal barrier of Caco-2 Cell monolayers through the expression of AMP-activated protein kinase-mediated Occludin, Claudin-1, and ZO-1. Biosci Biotechnol Biochem. 2015;79:130–137. doi: 10.1080/09168451.2014.951027. [DOI] [PubMed] [Google Scholar]
  • 241.Qiu J., Maekawa K., Kitamura Y., Miyata Y., Tanaka K., Tanaka T. Stimulation of glucose uptake by theasinensins through the AMP-activated protein kinase pathway in rat skeletal muscle cells. Biochem Pharmacol. 2014;87:344–351. doi: 10.1016/j.bcp.2013.10.029. [DOI] [PubMed] [Google Scholar]
  • 242.Bai T., Yang Y., Wu Y.L., Jiang S., Lee J.J., Lian L.H. Thymoquinone alleviates thioacetamide-induced hepatic fibrosis and inflammation by activating LKB1-AMPK signaling pathway in mice. Int Immunopharmacol. 2014;19:351–357. doi: 10.1016/j.intimp.2014.02.006. [DOI] [PubMed] [Google Scholar]
  • 243.Goto T., Teraminami A., Lee J.Y., Ohyama K., Funakoshi K., Kim Y.I. Tiliroside, a glycosidic flavonoid, ameliorates obesity-induced metabolic disorders via activation of adiponectin signaling followed by enhancement of fatty acid oxidation in liver and skeletal muscle in obese-diabetic mice. J Nutr Biochem. 2012;23:768–776. doi: 10.1016/j.jnutbio.2011.04.001. [DOI] [PubMed] [Google Scholar]
  • 244.Wu J.B., Kuo Y.H., Lin C.H., Ho H.Y., Shih C.C. Tormentic acid, a major component of suspension cells of Eriobotrya japonica, suppresses high-fat diet-induced diabetes and hyperlipidemia by glucose transporter 4 and AMP-activated protein kinase phosphorylation. J Agric Food Chem. 2014;62:10717–10726. doi: 10.1021/jf503334d. [DOI] [PubMed] [Google Scholar]
  • 245.Kopp C., Singh S.P., Regenhard P., Müller U., Sauerwein H., Mielenz M. Trans-cinnamic acid increases adiponectin and the phosphorylation of AMP-activated protein kinase through G-protein-coupled receptor signaling in 3T3-L1 adipocytes. Int J Mol Sci. 2014;15:2906–2915. doi: 10.3390/ijms15022906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Hu X., Wang S., Xu J., Wang D.B., Chen Y., Yang G.Z. Triterpenoid saponins from Stauntonia chinensis ameliorate insulin resistance via the AMP-activated protein kinase and IR/IRS-1/PI3K/AKT pathways in insulin-resistant HepG2 cells. Int J Mol Sci. 2014;15:10446–10458. doi: 10.3390/ijms150610446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Zheng Q.Y., Jin F.S., Yao C., Zhang T., Zhang G.H., Ai X. Ursolic acid-induced AMP-activated protein kinase (AMPK) activation contributes to growth inhibition and apoptosis in human bladder cancer T24 cells. Biochem Biophys Res Commun. 2012;419:741–747. doi: 10.1016/j.bbrc.2012.02.093. [DOI] [PubMed] [Google Scholar]
  • 248.Yang Y.B., Zhao Z.X., Liu Y.J., Kang X.J., Zhang H.S., Meng M. Suppression of oxidative stress and improvement of liver functions in mice by ursolic acid via LKB1-AMP-activated protein kinase signaling. J Gastroenterol Hepatol. 2015;30:609–618. doi: 10.1111/jgh.12723. [DOI] [PubMed] [Google Scholar]
  • 249.Lai C.S., Tsai M.L., Badmaev V., Jimenez M., Ho C.T., Pan M.H. Xanthigen suppresses preadipocyte differentiation and adipogenesis through down-regulation of PPARγ and C/EBPs and modulation of SIRT-1, AMPK, and FoxO pathways. J Agric Food Chem. 2012;60:1094–1101. doi: 10.1021/jf204862d. [DOI] [PubMed] [Google Scholar]
  • 250.Romero-Pérez A.I., Lamuela-Raventós R.M., Andrés-Lacueva C., de La Torre-Boronat M.C. Method for the quantitative extraction of resveratrol and piceid isomers in grape berry skins. Effect of powdery mildew on the stilbene content. J Agric Food Chem. 2001;49:210–215. doi: 10.1021/jf000745o. [DOI] [PubMed] [Google Scholar]
  • 251.Rencurel F., Foretz M., Kaufmann M.R., Stroka D., Looser R., Leclerc I. Stimulation of AMP-activated protein kinase is essential for the induction of drug metabolizing enzymes by phenobarbital in human and mouse liver. Mol Pharmacol. 2006;70:1925–1934. doi: 10.1124/mol.106.029421. [DOI] [PubMed] [Google Scholar]
  • 252.Rencurel F., Stenhouse A., Hawley S.A., Friedberg T., Hardie D.G., Sutherland C. AMP-activated protein kinase mediates phenobarbital induction of CYP2B gene expression in hepatocytes and a newly derived human hepatoma cell line. J Biol Chem. 2005;280:4367–4373. doi: 10.1074/jbc.M412711200. [DOI] [PubMed] [Google Scholar]

Articles from Acta Pharmaceutica Sinica. B are provided here courtesy of Elsevier

RESOURCES