Skip to main content
Cell Cycle logoLink to Cell Cycle
. 2011 Oct 15;10(20):3452–3460. doi: 10.4161/cc.10.20.17953

Bioenergy sensing in the brain

The role of AMP-activated protein kinase in neuronal metabolism, development and neurological diseases

Stephen Amato 1, Heng-Ye Man 1,
PMCID: PMC3266175  PMID: 22067656

Abstract

Bioenergy homeostasis constitutes one of the most crucial foundations upon which other cellular and organismal processes may be executed. AMP-activated protein kinase (AMPK) has been shown to be the key player in the regulation of energy metabolism, and thus is becoming the focus of research on obesity, diabetes and other metabolic disorders. However, its role in the brain, the most energy-consuming organ in our body, has only recently been studied and appreciated. Widely expressed in the brain, AMPK activity is tightly coupled to the energy status at both neuronal and whole-body levels. Importantly, AMPK signaling is intimately implicated in multiple aspects of brain development and function including neuronal proliferation, migration, morphogenesis and synaptic communication, as well as in pathological conditions such as neuronal cell death, energy depletion and neurodegenerative disorders.

Key words: AMPK, energy, neuron, brain, metabolism, glucose, neurodegenerative disease, cell death, neural development, polarization

Introduction

The intracellular environment is rarely in a state of static equilibrium, but instead exists in a state of constant energetic flux. Since bioenergy supply constitutes the very foundation for all forms of cellular activities, it is essential for cells to be equipped with a mechanism that couples activity and energy consumption to metabolism and energy supply. To meet this demand, eukaryotic cells have evolved AMP-activated protein kinase (AMPK), a protein capable of monitoring energy expenditure and efficiently regulating energy homeostasis.

AMPK monitors cellular energy status through sensitivity to the cellular AMP:ATP ratio, becoming active in response to metabolic stresses that decrease concentrations of ATP. Once activated, AMPK utilizes its serine/threonine kinase activity to increase the rate of cellular catabolism and simultaneously inhibit anabolic processes, resulting in a net increase in ATP production. The downstream pathways regulated by AMPK present an ever growing list that includes glycolysis, fatty acid oxidation and mTOR-mediated protein synthesis.13 However, AMPK's role is not limited to cell-autonomous energy regulation; many metabolic hormones, including leptin and adiponectin, function as upstream modulators of AMPK activity, allowing AMPK to participate in the maintenance of whole-body energy homeostasis. Additionally, the anti-diabetic drug metformin has also been observed to promote AMPK activity under a variety of experimental conditions.47

The importance of AMPK in general metabolic regulation has been appreciated for a long time, but its role in brain development and function has only recently been investigated. Given that the brain is the most energy-consuming organ in the body, it is logical to assume a crucial role of AMPK in neuronal energy regulation and brain activity. In this review, we will provide an overview on the general structure and regulation of AMPK before expanding on recent insights highlighting the role of the AMPK signaling cascade in the brain, including energy metabolism, neuroprotection/toxicity and neurodevelopment as well as the involvement of AMPK activity in certain neuropathological conditions.

AMPK Structure and Isoform Distribution

AMPK is a heterotrimeric protein complex that consists of α, β and γ subunits in equal stoichiometry. While the α subunit confers catalytic kinase activity, the β and γ subunits function in a regulatory capacity. The catalytic domain of the α subunit is located on the N-terminal region of the protein, while the C terminus is required for interaction with the β and γ subunits and complex association8 (Fig. 1). Variation between mammalian α isoforms is limited to non-catalytic regions, as the N-terminal catalytic core of α1 and α2 share 90% amino acid sequence identity, whereas the remaining protein displays only 61% homology.9 Also conserved within the α subunit is an autoinhibitory sequence (AIS) C-terminal relative to the kinase domain. The AIS sequence has been shown to repress kinase activity, as bacterially expressed constructs that contain both the kinase domain and the AIS sequence are roughly ten times less active than constructs that contain the kinase domain alone.10,11 Conservation within the β subunit is located to a central glycogen-binding domain (GBD) and a C-terminal region that functions as a scaffolding domain, allowing its association with both α and γ subunits.12 Mammalian γ subunits contain four tandem repeats named crystathionine β-synthase (CBS) domains, as the repeats were first identified in the enzyme crystahionine β-synthase.13 These domains function in tandem pairs, referred to as Bateman domains, binding to one molecule of either AMP or ATP in a mutually exclusive manner.1417

Figure 1.

Figure 1

Isoforms and splice variants of mammalian AMPK. The two α subunit isoforms each contain an N-terminal catalytic domain that shares 90% homology, including a threonine residue (T172) that is phosphorylated by upstream kinases for activation. While both isoforms exist within the brain, α2 is dominantly expressed in neurons. The two β subunits contain a glycogen-binding domain that is thought to play a role in the subcellular localization of AMPK in muscle cells and a C-terminal domain required for interaction with the α and γ subunits and complex formation. Three different isoforms exist for the γ subunit (γ1, γ2 and γ3), with γ2 and γ3 each possessing two splice variants (long and short forms) that vary in the length of the N-terminal domain (NTD). γ1 is the predominant isoform within the CNS, with γ3 being restricted to skeletal muscle.

Multiple isoforms exist for each of the three subunits, with both the α and β subunits having two isoforms (α1, α2, β1, β2), while three known isoforms exist for the γ subunit (γ1, γ2 and γ3).18 However, expression of γ3 is restricted to skeletal muscle.19 A study investigating the distribution of the various AMPK subunit isoforms in mice found that all three subunit isoforms exist in the CNS, but expression varies among different cell types. For instance, the α2 catalytic subunit is highly expressed in neurons compared with α1, which is expressed to a lesser degree in neurons. α subunits are not detected in glial cells at basal conditons, except in the activated astrocytes. The γ1 subunit is the dominant isoform expressed in neurons but is absent in astrocytes, while the levels of β1 and β2 subunit expression vary among different CNS cell types. Moreover, the expression of these isoforms within the brain is not static but, instead, displays developmental changes. It has been found that the mRNA levels for α2 and β2 subunits increase between embryonic days 10 and 14, whereas expression of α1, β1 and γ1 is consistent throughout ages E10 to P25.20 Currently, the basis for distinct spatial and temporal AMPK subunit expression remains unknown; however, higher affinity of the β2 for glycogen, compared with that of β1, has recently been revealed, possibly explaining the predominant expression of this isoform in muscle cells.21

Regulation of AMPK Activity

Basis for molecular activation.

The ability of the Bateman domains to bind either AMP or ATP exclusively, confers the sensitivity of AMPK to the cellular AMP:ATP ratio, and ultimately allows the protein to “sense” the energy level of the cell. More specifically, the binding of AMP to the γ subunit Bateman domains promotes activation of AMPK in three distinct ways.

First, the Bateman domains bind AMP in a cooperative manner, meaning binding of AMP to the first Bateman domain enables AMP binding to the second with greater affinity. Second, AMP binding to the γ subunit produces a conformational change in the AMPK complex, exposing threonine-172 (Thr172) in an activation loop of the α subunit. Phosphorylation of Thr172 by upstream kinases, referred to as AMPK kinases (AMPKKs), cause a roughly 50–100-fold increase in AMPK activity.22 Lastly, in addition to making AMPK a better substrate for AMPKKs, AMP binding also inhibits protein phosphatase-mediated dephosphorylation of Thr172.23 The combination of all three activation mechanisms enables AMPK to respond to very low levels of AMP, making it highly sensitive to slight fluctuations in cellular energy levels. Important to AMPK's role as an energy sensor, Bateman domains are also capable of binding ATP in a cooperative manner, although with much lower affinity than AMP, producing an antagonistic effect on AMPK activation.14 The inverse relationship between AMP and ATP binding in regard to AMPK activation creates a negative feedback loop, ensuring that AMPK activity decreases once cellular energy is recovered.

Regulation of AmpK by upstream kinases.

In addition to a rise in cellular AMP:ATP ratio and subsequent AMP binding, the ability of AMPK to regulate metabolic activity relies heavily upon Thr172 phosphorylation by upstream kinases. However, the identities of the kinases responsible for AMPK α subunit phosphorylation were unknown for decades following the initial discovery of AMPK. The first clue as to the identity of potential AMPKKs came from screening studies that identified multiple upstream kinases for the yeast homolog of AMPK, Snf1.2426 This information was used to identify two mammalian AMPKKs: the tumor suppressor LKB1 27,28 and the calmodulin-dependent protein kinase kinases, CaMKKα and CaMKKβ.2931

LKB1 was originally acknowledged as the tumor suppressor mutated in the genetically inherited susceptibility to human cancer, coined Peutz-Jeghers Syndrome.32 In regard to AMPK activation, studies performed in peripheral tissues demonstated that LKB1 is indeed necessary for phospho-activation of AMPK.27,33 Despite both LKB1 and AMPK being ubiquitously expressed in mammalian cells, there is evidence to suggest that AMPK may be acted upon by different AMPKKs in a tissue-specific manner. For instance, LKB1 has been demonstrated to be the major upstream activator of AMPK in muscle34 and liver cells;35 however, a study utilizing LKB1-knockouts found that LKB1 deficient neurons had similar levels of phosphorylated AMPK as compared with wild-type cells under normal physiological conditions.36 This observation suggests that another kinase may fulfill the role as the major AMPKK in the brain. To this end, Hawley et al. were able to show that in rat brain slices, intracellular increases in calcium following membrane depolarization resulted in a high degree of AMPK phosphorylation that was blocked by the CaMKK inhibitor STO-609. Because membrane depolarization did not affect the cellular AMP:ATP ratio, this study suggests that AMPK can be regulated in a Ca2+-dependent, AMP-independent manner.29 It will be interesting to know whether CaMKK is sufficient to phosphorylate neuronal AMPK during periods of energetic stress.

Ongoing research into identifying potential AMPKKs has revealed additional kinases in various tissues. The member of the mitogen-activated protein kinase kinase kinase family, TGFβ-activated kinase-1 (TAK1), was observed to phosphorylate AMPK in HeLa cells that lack LKB1 activity.37 Additionally, in cardiomyocytes, dominant-negative expression and gene knockout of TAK1 caused inhibition of AMPK T172 phosphorylation. TAK1 activity was found to be required in the activation of AMPK in response to Metformin, AICAR and ischemia.37,38 Although these studies make a strong point for TAK1 regulation of AMPK in peripheral tissues, it remains unclear whether TAK1 functions as a major regulator of AMPK in the brain.

AMPK Regulates Glucose Transport in Neurons

The brain constitutes only 2% of the body's weight but utilizes 50% of the total glucose supply. This can be attributed to the fact that neurons are the most metabolically demanding cells. But of more relevance, unlike peripheral tissues that utilize multiple nutrient sources for energy production, neurons use only glucose as fuel. The need for steady glucose uptake is compounded by the fact that neurons do not store glycogen and, therefore, must tightly couple energy demand to glucose influx in order to ensure a stable and continuous energy supply.39 In support of this, research has begun to emerge illustrating the relationship between AMPK activity and various energy-consuming neuronal activities.

In the brain, glutamate is the major neurotransmitter mediating most of the synaptic transmission. Multiple molecular events occurring during synaptic activation, such as sodium pump activity, receptor trafficking, cytoskeletal rearrangements, signaling and metabolic processes, make synaptic activity an energetically costly endeavor. It is thus conceivable for a crosstalk between glutamatergic excitation and energy mobilization. In line with this point, a study by Weisova et al. shows that cerebellar granule neurons had increased levels of phosphorylated AMPK in response to glutatmate stimulation, which was accompanied by a decrease in cellular ATP concentrations,40 explaining the basis for AMPK activation. Furthermore, glutamate excitation produced an increase in the surface expression of the glucose transporter GLUT3 that persisted for hours following the initial glutamate treatment. Importantly, increased GLUT3 surface expression was mediated by AMPK, as inhibition of AMPK, either through knockdown or pharmacological inhibition, blocked the GLUT3 response to glutamate. Interestingly, CaMKK inhibition had little effect on AMPK-mediated GLUT3 translocation, indicating AMPK activation by an alternative AMPKK during glutamate activation. Consistently, AMPK activity also translocates glucose transporters to the plasma membrane in skeletal and cardiac muscle during exercise;4144 these findings implicate AMPK as a crucial mediator coupling increased energy demands associated with neuronal activity and glucose uptake.

In peripheral tissues, insulin is a key signaling component in glucose uptake, while the regulation of neuronal glucose influx has traditionally been viewed as an insulin-insensitive process. However, there is evidence to suggest that both the insulin and insulin-like growth factor 1 (IGF1) signaling pathways are involved in glucose uptake in the brain.4547 For instance, suppressing endogenous insulin signaling in humans using somatostatin results in decreased glucose uptaking in the brain, as monitored by 18-fluorodeoxyglucose uptake and positron emission tomography (FDG-PET).48 Likewise, insulin treatment in cultured hippocampal neurons facilitates GLUT3 membrane translocation, which requires membrane depolarization for the final fusion of GLUT3-carrying vesicles with the plasma membrane to facilitate glucose uptake.49 Furthermore, IGF1 expression, which is closely related to insulin but shows an increased ability to cross the blood brain barrier50 and higher mRNA expression within the brain,51,52 was observed to parallel radiolabeled glucose uptake in mouse brains.46 In this same study, IGF1-null mouse brains showed reduced glucose uptake, implicating a role for IGF1 signaling in the regulation of glucose uptake within the brain.

Both insulin and IGF1 bind to receptor tyrosine kinases that associate with insulin receptor substrate (IRS),53 a known upstream activator of the PI3K/Akt signaling cascade.54 In peripheral cells, PI3K activation plays a significant role in glucose uptake55,56 and, therefore, may be involved in glucose influx within the brain as well. To this end, insulin increased GLUT4 translocation to the plasma membrane in human SH-SY5Y neuronal cell lines in a PI3K-dependent manner.57 Furthermore, in mouse brains, GLUT4 was localized to neuronal processes rich in both IGF1 and phosphorylated Akt, suggesting a physical association between these two proteins. Moreover, in Igf1-null brains, GLUT4 expression as well as Akt phosphorylation were reduced, suggesting that Igf1-induced Akt phosphorylation may play a role in translocation of GLUT4 to the neuronal membrane.46

Given that insulin/IGF1 participate in the regulation of intracellular transport of glucose, it is reasonable to hypothesize a connection between the energy sensor AMPK and the insulin/IGF cascade. In fact, AMPK is capable of phosphorylating IRS-1, the most upstream component of the insulin/IGF1 signaling cascade, in cell-free assays and mouse myoblast C2C12 cell lines in response to AICAR.58 In further support of this notion, our recent work showed that in both cultured rat embryonic hippocampal neurons and embryonic cortical brain slices, pharmacological activation of AMPK with the agonist AICAR resulted in a robust increase in the activity of the PI3K-Akt system.59 Importantly, increase in pAkt was blocked by inhibition of AMPK or PI3K activity, confirming a sequential activation of PI3K and Akt downstream of AMPK. Theses findings suggest that by activation of the PI3K/Akt cascade, probably via phosphorylation of IRS-1, AMPK relays energy-lacking signals to the glucose transport system to facilitate energy production.

AMPK Regulates Mitochondrial Function and Biogenesis

Mitochondrial function and biogenesis appear to be an important target of AMPK activation as an effort to increase cellular energy for extended periods of time. In fact, a major target of the AMPK cascade is the phosphorylation and subsequent inhibition of acetyl-CoA carboxylase (ACC),60,61 the enzyme responsible for converting acetyl-CoA to malonyl-CoA. By reducing malonyl-CoA production, AMPK facilitates CPT1-mediated long chain fatty acyl-CoA transport into the mitochondria, where the catabolic process of fatty acid β-oxidation can then occur.62

Skeletal muscles, much like neurons, are subject to high metabolic demands, as they are commonly required to perform multiple contractions over short periods of time. To maintain energy stores, muscle cells upregulate mitochondrial enzymes to compensate for chronic contraction and fatigue.63 More recently, experiments performed in skeletal muscle show that lasting contraction also results in elevated AMPK activity.64 Furthermore, chronic subcutaneous injection of AICAR into rat quadriceps increased mitochondrial enzyme cytochrome c, GLUT4 expression and ATP concentration.64 AMPK activity has also been linked to mitochondrial biogenesis, as mice expressing a dominant-negative AMPK mutant fail to increase their mitochondria mass in response to β-guanidinopropionic acid, a creatine analog that mimics endurance exercise and increases mitochondrial biogenesis in wild-type mice.65 Mechanistically, this was found to result from the ability of AMPK to activate PGC-1α, a transcriptional coactivator that, along with the transcription factor NRF-1, upregulates nuclear genes required for mitochondrial biogenesis.6568

In addition to muscle cells, recent studies show that mitochondrial biogenesis in neurons can also be regulated by AMPK. For instance, RT-PCR performed on Neuro2a cells treated with resveratrol, a polyphenol shown to activate AMPK in culture,69 revealed increased mRNA levels for the mitochondrial protein marker mitofusin 2 and two key regulators of mitochondrial biogenesis, PGC-1α and mitochondrial transcription factor A (mtTFA).69,70 Furthermore, the addition of the AMPK antagonist compound C, or expression of a dominant-negative AMPK mutant blocked the upregulation of these mitochondrial markers, supporting a role of AMPK in neuronal mitochondrial biogenesis. Another study showed that KCl depolarization of cultured rat visual cortical neurons activated AMPK and produced an increase in PGC-1α, NRF-1 and mtTFA levels and, furthermore, effectively increased ATP production.71 Addition of AICAR or resveratrol to neuron cultures also resulted in increases in PGC-1α and NRF-1 mRNA levels. Importantly, these changes were abolished by suppression of AMPK activity.

AMPK in Neurodevelopment

Proliferation and differentiation.

Although the role of energy homeostasis in mature brain functions has been extensively investigated, energy status in early brain development remains less well-understood. The complex cellular processes occurring during this time, including neuronal differentiation, migration, neurite growth and cellular polarization, consume a vast amount of energy and demand a secured and regulated energy supply.

In this regard, it has been found that loss of AMPK activity in Drosophila results in embryonic lethality, indicating an important role for AMPK activity in neural development.72 This study also suggests that AMPK is involved in maintaining genomic integrity during neural progenitor cell division, as a significant population of neuroblasts from AMPK-deficient larval brains displayed polyploidy.72 Additionally, another study utilizing Drosophila as a model system illustrated that loss of AMPK activity, via β-subunit knockout also results in extensive neurodegeneration shortly after neuronal differentiation.73

A study examining AMPK in the mammalian brain showed that knockout of AMPK β1 (AMPK β-/-) subunit in mice leads to severe loss of neurons and oligodendrocytes as well as abnormal astrocyte proliferation.74 Loss of AMPK functionality also produced proliferative defects and increased apoptosis within neural stem cell progenitors (NPCs), resulting in a 50% smaller brain than wild-type littermates. Retinoblastoma protein (Rb), known to be required in NPCs for normal brain development,75 was found to be directly phosphorylated by AMPK and showed hypophosphorylation in AMPK β-/- NPCs. Importantly, the misregulation of Rb was observed to prevent NPCs from exiting and reentering the cell cycle, thereby accounting for the decreased proliferation of AMPK β-/- NPCs.74 However, a study by Zhang et al. demonstrated that AICAR treatment and glucose deprivation caused cycle arrest in G0 phase and resulted in reduced proliferation of the immortalized mouse neural progenitor cell line, C17.2. This, again, was shown to result from the hypophosphorylation of Rb, an effect that was blocked by either compound C or expression of a dominant-negative AMPK.76 The differences observed may result from variations between the in vivo and cell line models utilized by these studies, but further investigation is required to solidify the effect AMPK activity has on proliferation within the CNS.

Polarization and migration.

Neuronal polarization constitutes the differentiation and elongation of minor neurites into multiple dendrites and a single axon. Because a neuron typically generates an axon of impressive size and complex geometry,77 its polarization entails extensive biosynthesis of protein and lipids, active transport of building materials to the axonal growth cone and extensive cytoskeleton rearrangements.78 These molecular processes must be sustained by a large amount of ATP consumption, suggesting potential sensitivity of neuronal polarization to cellular bioenergy status.79

Our recent study has demonstrated that when AMPK activity is pharmacologically enhanced to mimic energy-lacking conditions at the beginning of neuronal development, axon initiation and neuronal polarization are suppressed in both cultured embryonic hippocampal neurons and embryonic cortical brain slices, resulting in neurons that lack morphological polarization.59 This is concluded to result from AMPK-dependent disruption of PI3K enrichment at the neurite tip, previously shown to be a critical factor in axon initiation and polarization.80 Mechanistically, we find that direct phosphorylation of the light chain of the motor protein kinesin kif5 leads to the dissociation of the PI3K cargo from the motor complex and a failure of PI3K delivery to the neurite tip. Furthermore, we find that, although AMPK activity has a strong inhibitory effect on neuronal polarization, expression of a dominant-negative version of AMPK in both cultured neurons and brain slices has no effect on axon growth and polarization. In another study, knockout of both AMPKα1/α2 subunits in mice shows that AMPK activity is not required for cortical neurogenesis, neuronal migration or polarization under normal conditions.81 These findings point to a negligible role for basal AMPK activity during normal polarization, implying that AMPK may work to regulate neuronal polarization under pathological energy-lacking conditions. Indeed, a brief ischemia challenge during neuronal development results in inhibition of axon initiation and polarization (Amato, 2011). Further research is needed to determine whether neurons can resume axon growth after long periods of time, although short periods are not sufficient for recovery, as neurons remain unpolarized three days after removal of AMPK activators.59 Interestingly, LKB1, a major upstream activator of AMPK, has been shown to be required for polarization of cortical neurons in vivo.36,82 However, LKB1-deficient cortical neurons showed no change in phosphorylated or total AMPK levels, indicting that the effect of LKB1 in neuronal polarization is independent of AMPK activity.29

AMPK in Neuronal Viability

Many of the cellular insults that decrease cellular energy, such as ischemia and hypoxia, are known to induce apoptosis.83,84 It is thus not surprising to discover the involvement of AMPK activity in cell survival in both peripheral cells and the CNS. In C. elegans, activity of the LKB1/AMPK pathway is required for conservation of lipid reserves and thereby ensures survival of the larvae through maturation.85 Also, AMPK activity was observed to block TNFα-induced cardiomyocyte apoptosis that normally results from insults such as myocardial infarction and ischemia reperfusion,86 suggesting a neuroprotective role of the kinase.

In neurons, conflicting findings have been reported regarding the role of AMPK in cell death. AMPK activity appears to be both neuroprotective and pro-apoptotic, probably due to differences in nerve cell types, the nature of neural insults and the intensity and length of AMPK activation. For instance, pharmacological activation of AMPK via AICAR treatment effectively enhanced hippocampal neuron survival in response to glucose deprivation, chemical ischemia and glutamate exposure. Importantly, knockdown of the α1 and α2 subunits abolished the beneficial effects of AICAR, indicating direct involvement of AMPK activity.87 The neuroprotective effect may be mediated by neuronal inhibition. In response to ischemic insult, AMPK was observed to phosphorylate the R2 subunit of the GABAB receptor, a G protein-coupled receptor involved in post-synaptic hyperpolarization, resulting in enhanced receptor activity. AMPK-mediated upregulation in GABAB receptor activity has been shown to lead to post-synaptic hyperpolarization, reduced excitotoxicity and improved neuronal survival after chemical anoxia.88

Conversely, a study revealed that while AMPK activity significantly increased in response to ischemia, pharmacological inhibition of the kinase reduced stroke damage. Consistently, activation of AMPK through AICAR treatment intensified stroke damage in mice.89 Furthermore, the same study showed that knockout of AMPK α2 but not α1 subunit significantly reduced stroke-caused cell death, consistent with the results from AMPK inhibition.90

While knockout of AMPK activity in these experiments implicate direct involvement of AMPK, the wide-ranging effects AMPK imposes on cell survival begs further experimentation. It has been hypothesized that perhaps the difference between in vitro and in vivo findings stems from discrepancies in glucose supply, with glucose concentration in neuronal culture medium roughly ten times of that found at physiological conditions.91 Additionally, studies utilizing AICAR treatment showed that incubation time was also a factor in AMPK's effect on cell survival.92 Obviously, it is important to understand the molecular details underlying the different or opposite effects of AMPK on neuronal survival, so that a precise manipulation on AMPK signaling cascade can be designed for tentative clinical treatment.

AMPK in Neurodegenerative Diseases

AMPK is implicated in many metabolic diseases, including diabetes, obesity and cardiovascular disease. Recent studies have also revealed an important role of AMPK in neurodegenerative diseases, particularly Alzheimer disease (AD).93 AD is known to be associated with abnormalities in energy metabolism, including decreased glucose uptake and insulin sensitivity, compromised mitochondrial activities and impaired lipid metabolism.94,95 Hypometabolism and ATP depletion can cause partial membrane depolarization due to reduced sodium pump efficiency, which will potentiate NMDA receptor activity and eventually lead to neurotoxicity and cell death.96 Amyloid β (Aβ), the key molecule in AD pathogenesis, is a product of cleavage of its precursor protein APP by β-secretase (BACE1) and γ-secretase. It has been shown that in primary cortical neurons and N2a neuroblastoma cells, incubation with the AMPK activator metformin leads to an increase in both intracellular and extracellular Aβ generation. This effect is mediated by an elevated expression of BACE1 and abolished by AMPK inhibitor compound C, strongly indicating the direct role of AMPK in Aβ production.97 In contrast, a recent study showed that in cultured rat cortical neurons, activation of AMPK by AICAR treatment caused a significant reduction in Aβ. Consistently, AMPK α2 knockout led to increased Aβ production.98 Although possibly due to the use of a different AMPK activator or the intensity of AMPK activation, the exact reasons for the conflicting observations are not clear. The inhibitory effect of AMPK on amyloid production is supported by another study showing that activation of AMPK, either directly by AICAR treatment or indirectly via CaMKKβ following resveratrol incubation, lead to a reduction in Aβ levels.99 This finding illustrates AMPK signaling as the downstream event in resveratrol-induced Aβ downregulation.100 In addition, AMPK cascade also regulates Aβ degradation and removal from neurons.101

Hyperphosphorylaiton of microtubule-associated protein tau is another hallmark of AD. To date, more than 30 protein kinases have been implicated in tau phosphorylation, in which GSK3β (glycogen synthase kinase 3), Cdk5 (cyclin-dependent kinase 5), CK1 (casein kinase 1) and PKA play more important roles.102,103 In line with metabolic disregulation in AD, AMPK has recently been identified as a novel kinase targeting tau for protein phosphorylation. AMPK can phosphorylate tau at multiple sites, including Ser262, Ser356 and Ser396, reducing the interaction of tau with microtubules.104 Interestingly, this work indicates that Aβ itself can cause AMPK activation via regulation of NMDA receptor-dependent intracellular calcium homeostasis and CaMKKβ activity, indicating the existence of a complex, intertwined regulatory network in the development of AD.104 Furthermore, other AMPK family members, including MARK (microtubule-associated protein-regulating kinase/microtubule affinity-regulating kinase) and BRSK (brain-specific kinase) have also been shown to be involved in tau phosphorylation.104106

Although studies on the involvement of AMPK cascade in other neurodegenerative disorders are limited, its role does not seem limited to AD. For instance, in humans and mice with Huntington disease (HD), AMPK α1 subunit was selectively activated and enriched in the nucleus of striatal neurons. Overactivation of AMPK enhanced neuronal death and facilitated formation of Huntington aggregates. Importantly, suppression of AMPK activity or blocking its nuclear localization showed neuroprotective effects.107 AMPK activation is also observed in a paradigm of Parkinson disease (PD) induced by application of MPTP (1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine).108 Furthermore, AMPK may be implicated in neurodegenerative disorders through general cellular mechanisms. For example, dysfunction in protein degradation is a common feature shared by most neurodegenerative diseases, which leads to the formation of intracellular aggregates and neuronal toxicity. In supporting this notion, AMPK is a major player in the regulation of autophagic process in neurons via inhibition of mTOR pathway or activation of ATG1 enzyme.109,110

Concluding Remarks

As a master regulator of bioenergy status, AMPK has gained a growing amount of attention in its roles in numerous physiological and pathological systems. Its importance in neural tissue is especially exceptional, given that the brain is the most energy-demanding organ in the body and is therefore the most vulnerable to energy disregulation and depletion.111 AMPK is critically involved in multiple developmental stages and functionality of neurons, including migration, maturation, glucose uptake and metabolism and neuronal communication, including synaptic transmission112 and plasticity.113 At a system level, AMPK localized in energy-sensing neurons and circuits in the hypothalamus is responsible for the control of feeding behavior by incorporating peripheral energy-related signals.114117 It is worthy to note that at least 12 AMPK-related enzymes exist that show homology in the catalytic domain, constituting a AMPK-related kinase family that includes BRSK 1-2, NUAK 1-2, SIK 1-3, MARK 1-4 and MELK,118 but their functions remain largely unknown. We predict a rapid expansion in the investigation of AMPK and its related kinases, especially on their crucial roles in the formation and function of the nervous system

Acknowledgments

We thank Man lab members for helpful comments on the manuscript. The work in the laboratory of the authors is supported by National Institutes of Health Grant MH 079407 (H.Y.M.) and a NARSAD Young Investigator Award (H.Y.M.).

Abbreviations

ACC

acetyl-CoA carboxylase

AD

alzheimer disease

AICAR

5-aminoimidazole-4-carboxamide 1-β-D-ribofuranoside

AIS

autoinhibitory sequence

AMPKK

AMPK kinase

AMPK

AMP-activated protein kinase

BRSK

brain-specific kinase

CaMKK

calcium/calmodulin-dependent kinase kinase

CBS

crystathionine β-synthase

Cdk5

cyclin-dependent kinase 5

CK1

casein kinase 1

CNS

central nervous system

GLUT

glucose transporter

GSK

glycogen synthase kinase

HD

Huntington disease

IGF1

insulin-like growth factor 1

IRS

insulin receptor substrate

LKB1

liver kinase B1

MARK

microtubule-associated protein-regulating kinase/microtubule affinity-regulating kinase

mTOR

mammalian target of rapamycin

NTD

N-terminal domain

mtTFA

mitochondrial transcription factor A

NPC

neural progenitor cell

PI3K

phosphatidylinositol-3-kinases

TAK1

TGFβ-activated kinase-1

References

  • 1.Blagosklonny MV. Rapamycin and quasi-programmed aging: four years later. Cell Cycle. 2010;9:1859–1862. doi: 10.4161/cc.9.10.11872. [DOI] [PubMed] [Google Scholar]
  • 2.Blagosklonny MV. Calorie restriction: decelerating mTOR-driven aging from cells to organisms (including humans) Cell Cycle. 2010;9:683–688. doi: 10.4161/cc.9.4.10766. [DOI] [PubMed] [Google Scholar]
  • 3.Mounier R, Lantier L, Leclerc J, Sotiropoulos A, Foretz M, Viollet B. Antagonistic control of muscle cell size by AMPK and mTORC1. Cell Cycle. 2011;10:2640–2646. doi: 10.4161/cc.10.16.17102. [DOI] [PubMed] [Google Scholar]
  • 4.Anisimov VN, Egormin PA, Piskunova TS, Popovich IG, Tyndyk ML, Yurova MN, et al. Metformin extends life span of HER-2/neu transgenic mice and in combination with melatonin inhibits growth of transplantable tumors in vivo. Cell Cycle. 2010;9:188–197. doi: 10.4161/cc.9.1.10407. [DOI] [PubMed] [Google Scholar]
  • 5.Oliveras-Ferraros C, Vazquez-Martin A, Menendez JA. Genome-wide inhibitory impact of the AMPK activator metformin on [kinesins, tubulins, his-tones, auroras and polo-like kinases] M-phase cell cycle genes in human breast cancer cells. Cell Cycle. 2009;8:1633–1636. doi: 10.4161/cc.8.10.8406. [DOI] [PubMed] [Google Scholar]
  • 6.Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, et al. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest. 2001;108:1167–1174. doi: 10.1172/JCI13505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Zou MH, Kirkpatrick SS, Davis BJ, Nelson JS, Wiles WG, 4th, Schlattner U, et al. Activation of the AMP-activated protein kinase by the anti-diabetic drug metformin in vivo. Role of mitochondrial reactive nitrogen species. J Biol Chem. 2004;279:43940–43951. doi: 10.1074/jbc.M404421200. [DOI] [PubMed] [Google Scholar]
  • 8.Hudson ER, Pan DA, James J, Lucocq JM, Hawley SA, Green KA, et al. 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]
  • 9.Stapleton D, Mitchelhill KI, Gao G, Widmer J, Michell BJ, Teh T, et al. Mammalian AMP-activated protein kinase subfamily. J Biol Chem. 1996;271:611–614. doi: 10.1074/jbc.271.2.611. [DOI] [PubMed] [Google Scholar]
  • 10.Crute BE, Seefeld K, Gamble J, Kemp BE, Witters LA. Functional domains of the alpha1 catalytic subunit of the AMP-activated protein kinase. J Biol Chem. 1998;273:35347–35354. doi: 10.1074/jbc.273.52.35347. [DOI] [PubMed] [Google Scholar]
  • 11.Pang T, Xiong B, Li JY, Qiu BY, Jin GZ, Shen JK, et al. Conserved alpha-helix acts as autoinhibitory sequence in AMP-activated protein kinase alpha subunits. J Biol Chem. 2007;282:495–506. doi: 10.1074/jbc.M605790200. [DOI] [PubMed] [Google Scholar]
  • 12.Polekhina G, Gupta A, van Denderen BJ, Feil SC, Kemp BE, Stapleton D, et al. 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]
  • 13.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]
  • 14.Scott JW, Hawley SA, Green KA, Anis M, Stewart G, Scullion GA, et al. 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]
  • 15.Xiao B, Heath R, Saiu P, Leiper FC, Leone P, Jing C, et al. Structural basis for AMP binding to mammalian AMP-activated protein kinase. Nature. 2007;449:496–500. doi: 10.1038/nature06161. [DOI] [PubMed] [Google Scholar]
  • 16.Jin X, Townley R, Shapiro L. Structural insight into AMPK regulation: ADP comes into play. Structure. 2007;15:1285–1295. doi: 10.1016/j.str.2007.07.017. [DOI] [PubMed] [Google Scholar]
  • 17.Xiao B, Sanders MJ, Underwood E, Heath R, Mayer FV, Carmena D, et al. 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]
  • 18.Hardie DG. AMP-activated/SNF1 protein kinases: conserved guardians of cellular energy. Nat Rev Mol Cell Biol. 2007;8:774–785. doi: 10.1038/nrm2249. [DOI] [PubMed] [Google Scholar]
  • 19.Cheung PC, Salt IP, Davies SP, Hardie DG, Carling D. Characterization of AMP-activated protein kinase gamma-subunit isoforms and their role in AMP binding. Biochem J. 2000;346:659–669. doi: 10.1042/0264-6021:3460659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Turnley AM, Stapleton D, Mann RJ, Witters LA, Kemp BE, Bartlett PF. Cellular distribution and developmental expression of AMP-activated protein kinase isoforms in mouse central nervous system. J Neurochem. 1999;72:1707–1716. doi: 10.1046/j.1471-4159.1999.721707.x. [DOI] [PubMed] [Google Scholar]
  • 21.Koay A, Woodcroft B, Petrie EJ, Yue H, Emanuelle S, Bieri M, et al. AMPK beta 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]
  • 22.Hawley SA, Davison M, Woods A, Davies SP, Beri RK, Carling D, et al. Characterization of the AMP-activated protein kinase kinase from rat liver and identification of threonine 172 as the major site at which it phosphorylates AMP-activated protein kinase. J Biol Chem. 1996;271:27879–27887. doi: 10.1074/jbc.271.44.27879. [DOI] [PubMed] [Google Scholar]
  • 23.Davies SP, Helps NR, Cohen PT, Hardie DG. 5′-AMP inhibits dephosphorylation, as well as promoting phosphorylation, of the AMP-activated protein kinase. Studies using bacterially expressed human protein phosphatase-2Calpha and native bovine protein phosphatase-2AC. FEBS Lett. 1995;377:421–425. doi: 10.1016/0014-5793(95)01368-7. [DOI] [PubMed] [Google Scholar]
  • 24.Nath N, McCartney RR, Schmidt MC. Yeast Pak1 kinase associates with and activates Snf1. Mol Cell Biol. 2003;23:3909–3917. doi: 10.1128/MCB.23.11.3909-17.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Hong SP, Leiper FC, Woods A, Carling D, Carlson M. Activation of yeast Snf1 and mammalian AMP-activated protein kinase by upstream kinases. Proc Natl Acad Sci USA. 2003;100:8839–8843. doi: 10.1073/pnas.1533136100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Sutherland CM, Hawley SA, McCartney RR, Leech A, Stark MJ, Schmidt MC, et al. Elm1p is one of three upstream kinases for the Saccharomyces cerevisiae SNF1 complex. Curr Biol. 2003;13:1299–1305. doi: 10.1016/S0960-9822(03)00459-7. [DOI] [PubMed] [Google Scholar]
  • 27.Hawley SA, Boudeau J, Reid JL, Mustard KJ, Udd L, Makela TP, et al. Complexes between the LKB1 tumor suppressor, STRAD alpha/beta and MO25 alpha/beta 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]
  • 28.Woods A, Johnstone SR, Dickerson K, Leiper FC, Fryer LG, Neumann D, et al. 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]
  • 29.Hawley SA, Pan DA, Mustard KJ, Ross L, Bain J, Edelman AM, et al. Calmodulin-dependent protein kinase kinase-beta 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]
  • 30.Hurley RL, Anderson KA, Franzone JM, Kemp BE, Means AR, Witters LA. The Ca2+/calmodulin-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]
  • 31.Woods A, Dickerson K, Heath R, Hong SP, Momcilovic M, Johnstone SR, et al. Ca2+/calmodulin-dependent protein kinase kinase-beta 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]
  • 32.Hemminki A, Avizienyte E, Roth S, Loukola A, Aaltonen LA, Jarvinen H, et al. A serine/threonine kinase gene defective in Peutz-Jeghers syndrome. Duodecim. 1998;114:667–668. [PubMed] [Google Scholar]
  • 33.Shaw RJ, Kosmatka M, Bardeesy N, Hurley RL, Witters LA, DePinho RA, et al. The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc Natl Acad Sci USA. 2004;101:3329–3335. doi: 10.1073/pnas.0308061100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Sakamoto K, McCarthy A, Smith D, Green KA, Grahame Hardie D, Ashworth A, et al. 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]
  • 35.Shaw RJ, Lamia KA, Vasquez D, Koo SH, Bardeesy N, Depinho RA, et al. The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin. Science. 2005;310:1642–1646. doi: 10.1126/science.1120781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Barnes AP, Lilley BN, Pan YA, Plummer LJ, Powell AW, Raines AN, et al. LKB1 and SAD kinases define a pathway required for the polarization of cortical neurons. Cell. 2007;129:549–563. doi: 10.1016/j.cell.2007.03.025. [DOI] [PubMed] [Google Scholar]
  • 37.Momcilovic M, Hong SP, 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]
  • 38.Xie M, Zhang D, Dyck JR, Li Y, Zhang H, Morishima M, et al. A pivotal role for endogenous TGFbeta-activated kinase-1 in the LKB1/AMP-activated protein kinase energy-sensor pathway. Proc Natl Acad Sci USA. 2006;103:17378–17383. doi: 10.1073/pnas.0604708103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Vilchez D, Ros S, Cifuentes D, Pujadas L, Valles J, Garcia-Fojeda B, et al. Mechanism suppressing glycogen synthesis in neurons and its demise in progressive myoclonus epilepsy. Nat Neurosci. 2007;10:1407–1413. doi: 10.1038/nn1998. [DOI] [PubMed] [Google Scholar]
  • 40.Weisová P, Concannon CG, Devocelle M, Prehn JH, Ward MW. Regulation of glucose transporter 3 surface expression by the AMP-activated protein kinase mediates tolerance to glutamate excitation in neurons. J Neurosci. 2009;29:2997–3008. doi: 10.1523/JNEUROSCI.0354-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Fujii N, Jessen N, Goodyear LJ. AMP-activated protein kinase and the regulation of glucose transport. Am J Physiol Endocrinol Metab. 2006;291:867–877. doi: 10.1152/ajpendo.00207.2006. [DOI] [PubMed] [Google Scholar]
  • 42.Abbud W, Habinowski S, Zhang JZ, Kendrew J, Elkairi FS, Kemp BE, et al. Stimulation of AMP-activated protein kinase (AMPK) is associated with enhancement of Glut1-mediated glucose transport. Arch Biochem Biophys. 2000;380:347–352. doi: 10.1006/abbi.2000.1935. [DOI] [PubMed] [Google Scholar]
  • 43.Barnes K, Ingram JC, Porras OH, Barros LF, Hudson ER, Fryer LG, et al. Activation of GLUT1 by metabolic and osmotic stress: potential involvement of AMP-activated protein kinase (AMPK) J Cell Sci. 2002;115:2433–2442. doi: 10.1242/jcs.115.11.2433. [DOI] [PubMed] [Google Scholar]
  • 44.Cidad P, Almeida A, Bolanos JP. Inhibition of mitochondrial respiration by nitric oxide rapidly stimulates cytoprotective GLUT3-mediated glucose uptake through 5′-AMP-activated protein kinase. Biochem J. 2004;384:629–636. doi: 10.1042/BJ20040886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Hoyer S, Henneberg N, Knapp S, Lannert H, Martin E. Brain glucose metabolism is controlled by amplification and desensitization of the neuronal insulin receptor. Ann NY Acad Sci. 1996;777:374–379. doi: 10.1111/j.1749-6632.1996.tb34448.x. [DOI] [PubMed] [Google Scholar]
  • 46.Cheng CM, Reinhardt RR, Lee WH, Joncas G, Patel SC, Bondy CA. Insulin-like growth factor 1 regulates developing brain glucose metabolism. Proc Natl Acad Sci USA. 2000;97:10236–10241. doi: 10.1073/pnas.170008497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Bondy CA, Cheng CM. Signaling by insulin-like growth factor 1 in brain. Eur J Pharmacol. 2004;490:25–31. doi: 10.1016/j.ejphar.2004.02.042. [DOI] [PubMed] [Google Scholar]
  • 48.Bingham EM, Hopkins D, Smith D, Pernet A, Hallett W, Reed L, et al. The role of insulin in human brain glucose metabolism: an 18fluoro-deoxyglucose positron emission tomography study. Diabetes. 2002;51:3384–3390. doi: 10.2337/diabetes.51.12.3384. [DOI] [PubMed] [Google Scholar]
  • 49.Uemura E, Greenlee HW. Insulin regulates neuronal glucose uptake by promoting translocation of glucose transporter GLUT3. Exp Neurol. 2006;198:48–53. doi: 10.1016/j.expneurol.2005.10.035. [DOI] [PubMed] [Google Scholar]
  • 50.Reinhardt RR, Bondy CA. Insulin-like growth factors cross the blood-brain barrier. Endocrinology. 1994;135:1753–1761. doi: 10.1210/en.135.5.1753. [DOI] [PubMed] [Google Scholar]
  • 51.Bartlett GR. Phosphorus assay in column chromatography. J Biol Chem. 1959;234:466–468. [PubMed] [Google Scholar]
  • 52.Bondy CA. Transient IGF-I gene expression during the maturation of functionally related central projection neurons. J Neurosci. 1991;11:3442–3455. doi: 10.1523/JNEUROSCI.11-11-03442.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Myers M, Jr, Sun XJ, White MF. The IRS-1 signaling system. Trends Biochem Sci. 1994;19:289–293. doi: 10.1016/0968-0004(94)90007-8. [DOI] [PubMed] [Google Scholar]
  • 54.Saltiel AR, Pessin JE. Insulin signaling pathways in time and space. Trends Cell Biol. 2002;12:65–71. doi: 10.1016/S0962-8924(01)02207-3. [DOI] [PubMed] [Google Scholar]
  • 55.Summers SA, Birnbaum MJ. A role for the serine/threonine kinase, Akt, in insulin-stimulated glucose uptake. Biochem Soc Trans. 1997;25:981–988. doi: 10.1042/bst0250981. [DOI] [PubMed] [Google Scholar]
  • 56.Rose AJ, Richter EA. Skeletal muscle glucose uptake during exercise: how is it regulated? Physiology (Bethesda) 2005;20:260–270. doi: 10.1152/physiol.00012.2005. [DOI] [PubMed] [Google Scholar]
  • 57.Benomar Y, Naour N, Aubourg A, Bailleux V, Gertler A, Djiane J, et al. Insulin and leptin induce Glut4 plasma membrane translocation and glucose uptake in a human neuronal cell line by a phosphatidylinositol-3-kinase-dependent mechanism. Endocrinology. 2006;147:2550–2556. doi: 10.1210/en.2005-1464. [DOI] [PubMed] [Google Scholar]
  • 58.Jakobsen SN, Hardie DG, Morrice N, Tornqvist HE. 5′-AMP-activated protein kinase phosphorylates IRS-1 on Ser-789 in mouse C2C12 myotubes in response to 5-aminoimidazole-4-carboxamide riboside. J Biol Chem. 2001;276:46912–46916. doi: 10.1074/jbc.C100483200. [DOI] [PubMed] [Google Scholar]
  • 59.Amato S, Liu X, Zheng B, Cantley L, Rakic P, Man HY. AMP-activated protein kinase regulates neuronal polarization by interfering with PI 3-kinase localization. Science. 2011;332:247–251. doi: 10.1126/science.1201678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Winder WW, Hardie DG. Inactivation of acetyl-CoA carboxylase and activation of AMP-activated protein kinase in muscle during exercise. Am J Physiol. 1996;270:299–304. doi: 10.1152/ajpendo.1996.270.2.E299. [DOI] [PubMed] [Google Scholar]
  • 61.Winder WW, Wilson HA, Hardie DG, Rasmussen BB, Hutber CA, Call GB, et al. Phosphorylation of rat muscle acetyl-CoA carboxylase by AMP-activated protein kinase and protein kinase A. J Appl Physiol. 1997;82:219–225. doi: 10.1152/jappl.1997.82.1.219. [DOI] [PubMed] [Google Scholar]
  • 62.Kahn BB, Alquier T, Carling D, Hardie DG. AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab. 2005;1:15–25. doi: 10.1016/j.cmet.2004.12.003. [DOI] [PubMed] [Google Scholar]
  • 63.Holloszy JO. Biochemical adaptations in muscle. Effects of exercise on mitochondrial oxygen uptake and respiratory enzyme activity in skeletal muscle. J Biol Chem. 1967;242:2278–2282. [PubMed] [Google Scholar]
  • 64.Winder WW, Holmes BF, Rubink DS, Jensen EB, Chen M, Holloszy JO. Activation of AMP-activated protein kinase increases mitochondrial enzymes in skeletal muscle. J Appl Physiol. 2000;88:2219–2226. doi: 10.1152/jappl.2000.88.6.2219. [DOI] [PubMed] [Google Scholar]
  • 65.Zong H, Ren JM, Young LH, Pypaert M, Mu J, Birnbaum MJ, et al. AMP kinase is required for mitochondrial biogenesis in skeletal muscle in response to chronic energy deprivation. Proc Natl Acad Sci USA. 2002;99:15983–15987. doi: 10.1073/pnas.252625599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Jäger S, Handschin C, St-Pierre J, Spiegelman BM. AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1alpha. Proc Natl Acad Sci USA. 2007;104:12017–12022. doi: 10.1073/pnas.0705070104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Wareski P, Vaarmann A, Choubey V, Safiulina D, Liiv J, Kuum M, et al. PGC-1{alpha} and PGC-1{beta} regulate mitochondrial density in neurons. J Biol Chem. 2009;284:21379–21385. doi: 10.1074/jbc.M109.018911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Palacios OM, Carmona JJ, Michan S, Chen KY, Manabe Y, Ward JL, 3rd, et al. Diet and exercise signals regulate SIRT3 and activate AMPK and PGC1alpha in skeletal muscle. Aging (Albany NY) 2009;1:771–783. doi: 10.18632/aging.100075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Dasgupta B, Milbrandt J. Resveratrol stimulates AMP kinase activity in neurons. Proc Natl Acad Sci USA. 2007;104:7217–7222. doi: 10.1073/pnas.0610068104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Onyango IG, Lu J, Rodova M, Lezi E, Crafter AB, Swerdlow RH. Regulation of neuron mitochondrial biogenesis and relevance to brain health. Biochim Biophys Acta. 2010;1802:228–234. doi: 10.1016/j.bbadis.2009.07.014. [DOI] [PubMed] [Google Scholar]
  • 71.Yu L, Yang SJ. AMP-activated protein kinase mediates activity-dependent regulation of peroxisome proliferator-activated receptor gamma coactivator1alpha and nuclear respiratory factor 1 expression in rat visual cortical neurons. Neuroscience. 2010;169:23–38. doi: 10.1016/j.neuroscience.2010.04.063. [DOI] [PubMed] [Google Scholar]
  • 72.Lee JH, Koh H, Kim M, Kim Y, Lee SY, Karess RE, et al. Energy-dependent regulation of cell structure by AMP-activated protein kinase. Nature. 2007;447:1017–1020. doi: 10.1038/nature05828. [DOI] [PubMed] [Google Scholar]
  • 73.Spasić MR, Callaerts P, Norga KK. Drosophila alicorn is a neuronal maintenance factor protecting against activity-induced retinal degeneration. J Neurosci. 2008;28:6419–6429. doi: 10.1523/JNEUROSCI.1646-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Dasgupta B, Milbrandt J. AMP-activated protein kinase phosphorylates retinoblastoma protein to control mammalian brain development. Dev Cell. 2009;16:256–270. doi: 10.1016/j.devcel.2009.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.McLear JA, Garcia-Fresco G, Bhat MA, Van Dyke TA. In vivo inactivation of pRb, p107 and p130 in murine neuroprogenitor cells leads to major CNS developmental defects and high seizure rates. Mol Cell Neurosci. 2006;33:260–273. doi: 10.1016/j.mcn.2006.07.012. [DOI] [PubMed] [Google Scholar]
  • 76.Zang Y, Yu LF, Nan FJ, Feng LY, Li J. AMP-activated protein kinase is involved in neural stem cell growth suppression and cell cycle arrest by 5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside and glucose deprivation by downregulating phospho-retinoblastoma protein and cyclin D. J Biol Chem. 2009;284:6175–6184. doi: 10.1074/jbc.M806887200. [DOI] [PubMed] [Google Scholar]
  • 77.Ropireddy D, Scorcioni R, Lasher B, Buzsaki G, Ascoli GA. Axonal morphometry of hippocampal pyramidal neurons semi-automatically reconstructed after in vivo labeling in different CA3 locations. Brain Struct Funct. 2011;216:1–15. doi: 10.1007/s00429-010-0291-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Arimura N, Kaibuchi K. Neuronal polarity: from extracellular signals to intracellular mechanisms. Nat Rev Neurosci. 2007;8:194–205. doi: 10.1038/nrn2056. [DOI] [PubMed] [Google Scholar]
  • 79.Amato S. AMPK links cellular bioenegy to the decision making of axon initiation in neurons. Cell Logist. 2011;1:103–105. doi: 10.4161/cl.1.3.16815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Shi SH, Jan LY, Jan YN. Hippocampal neuronal polarity specified by spatially localized mPar3/mPar6 and PI 3-kinase activity. Cell. 2003;112:63–75. doi: 10.1016/S0092-8674(02)01249-7. [DOI] [PubMed] [Google Scholar]
  • 81.Williams T, Courchet J, Viollet B, Brenman JE, Polleux F. AMP-activated protein kinase (AMPK) activity is not required for neuronal development but regulates axogenesis during metabolic stress. Proc Natl Acad Sci USA. 2011;108:5849–5854. doi: 10.1073/pnas.1013660108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Shelly M, Cancedda L, Heilshorn S, Sumbre G, Poo MM. LKB1/STRAD promotes axon initiation during neuronal polarization. Cell. 2007;129:565–577. doi: 10.1016/j.cell.2007.04.012. [DOI] [PubMed] [Google Scholar]
  • 83.Guo MF, Yu JZ, Ma CG. Mechanisms related to neuron injury and death in cerebral hypoxic ischaemia. Folia Neuropathol. 2011;49:78–87. [PubMed] [Google Scholar]
  • 84.Northington FJ, Chavez-Valdez R, Martin LJ. Neuronal cell death in neonatal hypoxia-ischemia. Ann Neurol. 2011;69:743–758. doi: 10.1002/ana.22419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Narbonne P, Roy R. Caenorhabditis elegans dauers need LKB1/AMPK to ration lipid reserves and ensure long-term survival. Nature. 2009;457:210–214. doi: 10.1038/nature07536. [DOI] [PubMed] [Google Scholar]
  • 86.Kewalramani G, Puthanveetil P, Wang F, Kim MS, Deppe S, Abrahani A, et al. AMP-activated protein kinase confers protection against TNF{alpha}-induced cardiac cell death. Cardiovasc Res. 2009;84:42–53. doi: 10.1093/cvr/cvp166. [DOI] [PubMed] [Google Scholar]
  • 87.Culmsee C, Monnig J, Kemp BE, Mattson MP. AMP-activated protein kinase is highly expressed in neurons in the developing rat brain and promotes neuronal survival following glucose deprivation. J Mol Neurosci. 2001;17:45–58. doi: 10.1385/JMN:17:1:45. [DOI] [PubMed] [Google Scholar]
  • 88.Kuramoto N, Wilkins ME, Fairfax BP, Revilla-Sanchez R, Terunuma M, Tamaki K, et al. Phospho-dependent functional modulation of GABA(B) receptors by the metabolic sensor AMP-dependent protein kinase. Neuron. 2007;53:233–247. doi: 10.1016/j.neuron.2006.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.McCullough LD, Zeng Z, Li H, Landree LE, McFadden J, Ronnett GV. Pharmacological inhibition of AMP-activated protein kinase provides neuroprotection in stroke. J Biol Chem. 2005;280:20493–20502. doi: 10.1074/jbc.M409985200. [DOI] [PubMed] [Google Scholar]
  • 90.Li J, Zeng Z, Viollet B, Ronnett GV, McCullough LD. Neuroprotective effects of adenosine monophosphateactivated protein kinase inhibition and gene deletion in stroke. Stroke. 2007;38:2992–2999. doi: 10.1161/STROKEAHA.107.490904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Ronnett GV, Ramamurthy S, Kleman AM, Landree LE, Aja S. AMPK in the brain: its roles in energy balance and neuroprotection. J Neurochem. 2009;109:17–23. doi: 10.1111/j.1471-4159.2009.05916.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Garcia-Gil M, Pesi R, Perna S, Allegrini S, Giannecchini M, Camici M, et al. 5′-aminoimidazole-4-carboxamide riboside induces apoptosis in human neuroblastoma cells. Neuroscience. 2003;117:811–820. doi: 10.1016/S0306-4522(02)00836-9. [DOI] [PubMed] [Google Scholar]
  • 93.Salminen A, Kaarniranta K, Haapasalo A, Soininen H, Hiltunen M. AMP-activated protein kinase: a potential player in Alzheimer's disease. J Neurochem. 2011;118:460–474. doi: 10.1111/j.1471-4159.2011.07331.x. [DOI] [PubMed] [Google Scholar]
  • 94.Ferreira IL, Resende R, Ferreiro E, Rego AC, Pereira CF. Multiple defects in energy metabolism in Alzheimer's disease. Curr Drug Targets. 2010;11:1193–1206. doi: 10.2174/1389450111007011193. [DOI] [PubMed] [Google Scholar]
  • 95.Hoyer S. Brain glucose and energy metabolism abnormalities in sporadic Alzheimer disease. Causes and consequences: an update. Exp Gerontol. 2000;35:1363–1372. doi: 10.1016/S0531-5565(00)00156-X. [DOI] [PubMed] [Google Scholar]
  • 96.Beal MF. Does impairment of energy metabolism result in excitotoxic neuronal death in neurodegenerative illnesses? Ann Neurol. 1992;31:119–130. doi: 10.1002/ana.410310202. [DOI] [PubMed] [Google Scholar]
  • 97.Chen Y, Zhou K, Wang R, Liu Y, Kwak YD, Ma T, et al. Antidiabetic drug metformin (GlucophageR) increases biogenesis of Alzheimer's amyloid peptides via upregulating BACE1 transcription. Proc Natl Acad Sci USA. 2009;106:3907–3912. doi: 10.1073/pnas.0807991106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Won JS, Im YB, Kim J, Singh AK, Singh I. Involvement of AMP-activated-protein-kinase (AMPK) in neuronal amyloidogenesis. Biochem Biophys Res Commun. 2010;399:487–491. doi: 10.1016/j.bbrc.2010.07.081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Vingtdeux V, Giliberto L, Zhao H, Chandakkar P, Wu Q, Simon JE, et al. AMP-activated protein kinase signaling activation by resveratrol modulates amyloid-beta peptide metabolism. J Biol Chem. 2010;285:9100–9113. doi: 10.1074/jbc.M109.060061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Marambaud P, Zhao H, Davies P. Resveratrol promotes clearance of Alzheimer's disease amyloidbeta peptides. J Biol Chem. 2005;280:37377–37382. doi: 10.1074/jbc.M508246200. [DOI] [PubMed] [Google Scholar]
  • 101.Vingtdeux V, Chandakkar P, Zhao H, d'Abramo C, Davies P, Marambaud P. Novel synthetic small-molecule activators of AMPK as enhancers of autophagy and amyloid-beta peptide degradation. FASEB J. 2011;25:219–231. doi: 10.1096/fj.10-167361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Hanger DP, Anderton BH, Noble W. Tau phosphorylation: the therapeutic challenge for neurodegenerative disease. Trends Mol Med. 2009;15:112–119. doi: 10.1016/j.molmed.2009.01.003. [DOI] [PubMed] [Google Scholar]
  • 103.Johnson GV, Stoothoff WH. Tau phosphorylation in neuronal cell function and dysfunction. J Cell Sci. 2004;117:5721–5729. doi: 10.1242/jcs.01558. [DOI] [PubMed] [Google Scholar]
  • 104.Thornton C, Bright NJ, Sastre M, Muckett PJ, Carling D. AMP-activated protein kinase (AMPK) is a tau kinase, activated in response to amyloid beta-peptide exposure. Biochem J. 2011;434:503–512. doi: 10.1042/BJ20101485. [DOI] [PubMed] [Google Scholar]
  • 105.Drewes G, Ebneth A, Preuss U, Mandelkow EM, Mandelkow E. MARK, a novel family of protein kinases that phosphorylate microtubule-associated proteins and trigger microtubule disruption. Cell. 1997;89:297–308. doi: 10.1016/S0092-8674(00)80208-1. [DOI] [PubMed] [Google Scholar]
  • 106.Wang JW, Imai Y, Lu B. Activation of PAR-1 kinase and stimulation of tau phosphorylation by diverse signals require the tumor suppressor protein LKB1. J Neurosci. 2007;27:574–581. doi: 10.1523/JNEUROSCI.5094-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Ju TC, Chen HM, Lin JT, Chang CP, Chang WC, Kang JJ, et al. Nuclear translocation of AMPK{alpha}1 potentiates striatal neurodegeneration in Huntington's disease. J Cell Biol. 2011;194:209–227. doi: 10.1083/jcb.201105010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Choi JS, Park C, Jeong JW. AMP-activated protein kinase is activated in Parkinson's disease models mediated by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Biochem Biophys Res Commun. 2010;391:147–151. doi: 10.1016/j.bbrc.2009.11.022. [DOI] [PubMed] [Google Scholar]
  • 109.Egan DF, Shackelford DB, Mihaylova MM, Gelino S, Kohnz RA, Mair W, et al. Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science. 2011;331:456–461. doi: 10.1126/science.1196371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Kim J, Kundu M, Viollet B, Guan KL. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol. 2011;13:132–141. doi: 10.1038/ncb2152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Pellerin L, Magistretti PJ. How to balance the brain energy budget while spending glucose differently. J Physiol. 2003;546:325. doi: 10.1113/jphysiol.2002.035105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Dhar SS, Wong-Riley MT. Coupling of energy metabolism and synaptic transmission at the transcriptional level: role of nuclear respiratory factor 1 in regulating both cytochrome c oxidase and NMDA glutamate receptor subunit genes. J Neurosci. 2009;29:483–492. doi: 10.1523/JNEUROSCI.3704-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Potter WB, O'Riordan KJ, Barnett D, Osting SM, Wagoner M, Burger C, et al. Metabolic regulation of neuronal plasticity by the energy sensor AMPK. PLoS ONE. 2010;5:8996. doi: 10.1371/journal.pone.0008996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Andersson U, Filipsson K, Abbott CR, Woods A, Smith K, Bloom SR, et al. AMP-activated protein kinase plays a role in the control of food intake. J Biol Chem. 2004;279:12005–12008. doi: 10.1074/jbc.C300557200. [DOI] [PubMed] [Google Scholar]
  • 115.Lim CT, Kola B, Korbonits M. AMPK as a mediator of hormonal signalling. J Mol Endocrinol. 2010;44:87–97. doi: 10.1677/JME-09-0063. [DOI] [PubMed] [Google Scholar]
  • 116.Minokoshi Y, Shiuchi T, Lee S, Suzuki A, Okamoto S. Role of hypothalamic AMP-kinase in food intake regulation. Nutrition. 2008;24:786–790. doi: 10.1016/j.nut.2008.06.002. [DOI] [PubMed] [Google Scholar]
  • 117.Ramamurthy S, Ronnett GV. Developing a head for energy sensing: AMP-activated protein kinase as a multifunctional metabolic sensor in the brain. J Physiol. 2006;574:85–93. doi: 10.1113/jphysiol.2006.110122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Bright NJ, Thornton C, Carling D. The regulation and function of mammalian AMPK-related kinases. Acta Physiol (Oxf) 2009;196:15–26. doi: 10.1111/j.1748-16.2009.01971.x. [DOI] [PubMed] [Google Scholar]

Articles from Cell Cycle are provided here courtesy of Taylor & Francis

RESOURCES