Abstract
The AMP-activated protein kinase α (AMPKα) subunit is the catalytic subunit in the AMPK complex, playing a crucial role in AMPK activation. It has two isoforms: AMPKα1 and AMPKα2. Emerging evidence suggests that the AMPKα subunit exhibits subtype-specific effects in Alzheimer's disease (AD). This review discusses the role of the AMPKα subunit in the pathogenesis of AD, including its impact on β-amyloid (Aβ) pathology, Tau pathology, metabolic disorders, inflammation, mitochondrial dysfunction, inflammasome and pyroptosis. Additionally, it reviews the distinct roles of its isoforms, AMPKα1 and AMPKα2, in AD, which may provide more precise targets for future drug development in AD.
Keywords: Alzheimer's disease, AMPKα subunit, AMPKα isoforms, Subtype specificity, Target precision
1. Introduction
Alzheimer's disease (AD) is the most common type of dementia. With the exacerbation of population aging, the incidence, prevalence, and mortality of AD are increasing, imposing a significant economic burden on families and society, making it a crucial public health issue [1,2]. Clinical manifestations of AD include decline in learning and memory abilities, cognitive impairment, behavioral abnormalities, and social dysfunction. Its typical pathological features include the formation of senile plaques (SPs) composed of β-amyloid (Aβ) outside cells, neurofibrillary tangles formed by excessive phosphorylation of tau protein inside cells, and degeneration and loss of neurons and synapses [2]. However, the etiology and pathogenesis of AD are not fully understood, and there are currently no effective interventions to cure or slow down the progression of AD. The brain, as one of the organs with high energy consumption, is vulnerable to disturbances in energy metabolism, with ample evidence indicating that reduced energy metabolism is an early and consistent feature of AD [3]. AMP-activated protein kinase (AMPK), as a cellular energy sensor, plays a significant role in the pathophysiology of AD [[4], [5], [6]] and may be associated with its pathogenesis [7], being considered a potential therapeutic target for AD.
AMPK is a sensor of cellular energy and nutrient status, activated under low energy conditions to maintain cellular homeostasis [8]. In eukaryotes, it is almost universally expressed as a heterotrimeric complex consisting of catalytic subunits (α), scaffolding subunits (β), and regulatory subunits (γ), in a 1:1:1 ratio [9]. AMPK is mainly activated through two mechanisms: (1) one is the AMP/ADP-dependent energy stress signal: in the absence of AMP, the autoinhibitory domain of AMPKα keeps the kinase in an inactive conformation; when the cellular AMP:ATP ratio increases, AMP binds to the γ subunit, leading to a conformational change in the α subunit or activation of AMPK by preventing phosphatases from dephosphorylating the Thr172 site [10]. Additionally, an increase in AMP levels can activate LKB1 [11], calcium/calmodulin-dependent protein kinase kinase (CaMKKβ) [12], or transforming growth factor β-activated kinase 1 (TAK1) [13], leading to phosphorylation of the α subunit Thr172 site and thereby activating AMPK. (2) Another non-AMP/ADP-dependent pathway involves the sensing of various nutrients: (a) Glucose: Activation of AMPK occurs through the sensing of fructose-1,6-bisphosphate (FBP) by aldolase, in the absence of which AMPK is activated [14]. Since the discovery of this pathway, glucose deprivation has been widely used as an effective method to activate AMPK in cultured cells [15]. The possible mechanism involved is that glucose deprivation leads to a conformational change in the v-ATPase regulatory complex, thereby activating AMPK [16]; (b) Fatty acids: Research on the allosteric drug and metabolite site ligands has shown that long-chain fatty acyl-CoAs (LCFA-CoAs) are natural ligands of AMPK, and their binding can activate AMPK [17]; (c) Glycogen: The AMPKβ subunit contains a glycogen-binding site on the β-carbohydrate binding module (β-CBM), which can stabilize AMPK in vivo [[18], [19], [20], [21]]. Additionally, AMPK activation also occurs in response to nuclear DNA and lysosomal damage, as well as ionizing radiation [[22], [23], [24], [25], [26], [27]]. Once activated, AMPK phosphorylates proteins involved in catabolic pathways to increase ATP synthesis, while phosphorylating or inactivating proteins involved in anabolic pathways. The dysregulation of AMPK signaling in AD has complex effects. For example, AMPK can promote the phosphorylation of Tau protein and inhibit Tau phosphorylation or aggregation by inhibiting glycogen synthase kinase 3β (GSK3β) or activating recombinant Sirtuin 1 (SIRT1) mediated Tau deacetylation [28,29]. AMPK activation can increase Aβ through endoplasmic reticulum stress [30] or upregulation of β-secretase (BACE1) [31], as well as enhance autophagy to increase Aβ clearance [32]. Activation of AMPK by metformin alleviates mitochondrial damage and improves pathology and cognitive function in AD models [33,34]; however, AMPK inhibitors correct the toxic effects of Aβ on synaptic function [4,5]. These controversies highlight the complexity of AMPK's role in AD and the complexity of AMPK itself. The α, β, and γ subunits of AMPK have multiple subtypes, and the activation of different subtypes may be the reason for the complexity of AMPK action [35]. Among them, the AMPKα subunit is considered a key factor determining AMPK activity [36] and plays a crucial role in regulating Aβ production [37] and aggregation [38], Tau phosphorylation [39] and aggregation [38], metabolic disorders [40], inflammation [41], mitochondrial dysfunction [42] and apoptosis [43] in AD. Recent studies have discovered subtype-specific roles of the AMPKα subunit in the pathogenesis of AD [44,45]. For instance, inhibiting AMPKα1 effectively rescues synaptic defects and memory loss in AD model mice [46], while AMPKα2 is associated with Aβ load in AD model mice [47]. Therefore, elucidating the role of the AMPKα subunit in AD may provide more precise therapeutic targets for AD treatment. This article will review the research progress on the role of the AMPKα subunit in the pathogenesis of AD.
2. AMPKα subunit and its isoforms
AMPKα subunit is the catalytic subunit of AMPK, with its N-terminus containing a traditional serine/threonine kinase domain, followed by an auto-inhibitory domain (AID), and then an extended "linker peptide" that connects the AID to the C-terminal domain (α-CTD) [48]. The α-CTD is a crucial structure for the interaction of the α subunit with the β and γ subunits [49]. The AMPKα subunit plays a significant role in AMPK activation, primarily through two general pathways: 1) Binding of AMP to the γ subunit domain causes a conformational change in the α subunit, activating AMPK; 2) Increased AMP levels activate the CaMKKβ [12], LKB1 [11] and TAK1 [13], leading to the phosphorylation of a serine residue in the N-terminal kinase domain activation segment of the α subunit (usually referred to as Thr-172 due to its position in the rat gene sequence) [50], thereby activating AMPK [[51], [52], [53]]. Therefore, the AMPKα subunit is a key factor determining the activity of AMPK [36].
The AMPKα subunit currently known has two subtypes: AMPKα1 and AMPKα2, encoded by the PRKAA1 gene on chromosome 5 and the PRKAA2 gene on chromosome 1, respectively [54]. The α1 and α2 subtypes exhibit tissue specificity. The α1 subtype is strongly expressed in embryos, but its expression decreases during development [55]. In adults, the expression of the α1 subtype in the liver is 250 times that of the α2 subtype [56]. In skeletal muscle and cardiac tissue, the α2 subtype is the main catalytic subtype and plays a major role in glucose uptake in skeletal muscle and energy metabolism in the heart [57,58]. In the central nervous system, both the α1 and α2 subtypes have high levels in the embryonic hippocampus and are present in neurons throughout the adult brain [55]. Specifically, the α1 subtype is mainly expressed in astrocytes, while the α2 subtype is the primary catalytic unit expressed in the adult brain and spinal cord, with the highest expression found in cortical and hippocampal neurons as well as Purkinje cells in the cerebellum [59]. Studies on the subcellular distribution of the α2 subtype at the central cell level suggest that in neurons, it is mainly located in the cell nucleus, while in glial cells, it is primarily found in the cytoplasm [59]. Recent evidence has revealed the potential roles of AMPKα subtypes in both neurophysiology and neuropathophysiology [6]. For instance, downregulation of the AMPKα1 subtype promotes oxidative stress in astrocytes [60]. Knockout of the AMPKα1 subtype in mice results in severe demyelination and inflammatory reactions in the brain and spinal cord [61]. Activation of the AMPKα1 subtype significantly inhibits the nuclear translocation of nuclear factor-k-gene binding (NF-κB) and the expression of pro-inflammatory factors in BV2 microglial cells [62]. These findings indicate a specific role of the AMPKα1 subtype in maintaining central oxidative balance, myelin formation, and anti-inflammatory functions. Similarly, the AMPKα2 subtype also exhibits specific functions in the central nervous system. For example, brain-specific inhibition of the AMPKα2 subtype leads to cognitive impairments, highlighting the importance of the AMPKα2 subtype in maintaining normal cognitive function [63]. Enzyme assays in the hypothalamic region of rodents suggest that appetite-suppressing hormones (such as the fat-derived hormone leptin) inhibit the AMPKα2 subtype [64], indicating a relationship between the AMPKα2 subtype and appetite control in the hypothalamus. In mice with the AMPKα2 gene knocked out, the expression of catecholamines increases [65], suggesting a link between the AMPKα2 subtype and the regulation of sympathetic nervous system activity. However, the exact role of AMPKα subtypes in the central nervous system remains incompletely understood at present.
One of the established downstream effects of AMPK is the regulation of protein synthesis or mRNA translation. AMPK can regulate protein synthesis through at least two classical mechanisms: eukaryotic elongation factor 2 kinase (eEF2K) and mammalian target of rapamycin complex 1 (mTORC1) signaling cascade targets. In brief, AMPK phosphorylates and activates eEF2K, leading to the phosphorylation of the translation factor eEF2, thereby inhibiting general protein synthesis. Additionally, AMPK inhibits (directly or indirectly) the mTORC1 signaling pathway, whose activation promotes the synthesis of translational machinery and cap-dependent translation initiation [54]. Several studies have shown that the AMPKα subunit can regulate changes in the mTOR-induced autophagy pathway [[66], [67], [68]], lipid metabolism [69,70], and transcriptomics show that knocking down the AMPKα subunit affects gene expression involved in lipid and carbohydrate metabolism as well as insulin/insulin-like growth factor signaling (IIS) [71]. However, it is not fully understood how AMPK subunits or isoforms affect the translation of specific proteins in the brain. Proteomic analysis of the hippocampus of mice with specific knockout of AMPKα1 and α2 isoforms has revealed the identities of proteins sensitive to AMPKα1 or α2 isoforms [72]. In the hippocampus of mice with knockout of AMPKα1 or AMPKα2 isoforms, upregulation of proteins related to binding, catalytic activity, structural molecular activity, and transporter protein activity was detected, with proteins such as protein kinase C in neuronal protein 2 and protein D2 containing an EF-hand domain being upregulated in both hippocampi. Conversely, downregulated proteins showed some differences between the two. In the hippocampus of AMPKα1 knockout mice, the downregulated proteins were classified into three categories: binding, catalytic activity, and translation regulatory activity, while in AMPKα2 knockout mice, they were classified into five categories: binding, catalytic activity, structural molecular activity, translation regulatory activity, and transporter protein activity [72]. It is worth noting that the proteasome subunit alpha type-7 was upregulated in the hippocampus of AMPKα2 knockout mice, and the protein Cullin-3, which was downregulated in the hippocampi of both AMPKα1 and α2 knockout mice, is involved in lysosomal protein hydrolysis and the ubiquitin-proteasome pathway, both of which are dysregulated in AD [72,73]. However, the exact impact of AMPKα subunits on the pathogenesis of AD is not yet fully understood.
3. AMPKα subunit and AD
The AMPKα subunit is correlated with different pathogenic mechanisms in AD (including Aβ pathology, Tau pathology, metabolic disorders, inflammation, mitochondrial dysfunction, and pyroptosis). Similarly, as our understanding of the AMPKα subunit deepens, it becomes increasingly clear that the AMPKα subunit plays a significant role in the onset and progression of AD. Here, we outline the potential roles and mechanisms of the AMPKα subunit in various pathogenic mechanisms of AD.
3.1. AMPKα subunit and Aβ
Aβ is the main component of SPs formed by the cleavage of amyloid precursor protein (APP) by BACE1 and γ-secretase [74]. The formation of Aβ in SPs is considered a key pathological event in AD [75]. Efforts have been made to explore methods to alleviate Aβ pathology to mitigate AD symptoms, including reducing Aβ production [[76], [77], [78]], inhibiting Aβ aggregation [[79], [80], [81]], and enhancing Aβ clearance [78,82,83]. The AMPKα subunit regulates the activation of AMPK, which can modulate cholesterol and sphingolipid metabolism, Cholesterol and sphingolipids are involved in the regulation of APP processing and Aβ production [84]. In neurons with AMPKα2 knockout, increased Aβ production suggests that activation of the AMPKα subunit reduces Aβ production by regulating the generation of APP [4]. Studies have shown that upregulation of AMPKα subunit can inhibit Aβ expression in the mouse brain [37]. Additionally, the phosphorylation of AMPKα has been found to inhibit Aβ aggregation [38]. Autophagy plays a neuroprotective role in AD by mediating the degradation of Aβ. The AMPKα subunit can inhibit mTOR to enhance autophagy, reducing Aβ deposition and exerting a neuroprotective effect [85]. While these studies demonstrate the potential of the AMPKα subunit in regulating Aβ pathology, further research, including clinical trials, is needed to validate its feasibility as a therapeutic target for AD. Nevertheless, experimental data support the promising role of the AMPKα subunit in regulating Aβ pathology and potentially alleviating the progression of AD.
3.2. AMPKα subunit and Tau
Tau is a microtubule-associated protein that stabilizes neuronal structures. However, in AD, Tau undergoes abnormal modifications, such as hyperphosphorylation, leading to the formation of neurofibrillary tangles within neurons [86]. Previous studies have shown that at 8 months of age, APP/PS1 transgenic mice exhibit hyperphosphorylated Tau-positive neuroinflammatory structures near amyloid plaques in the brain [87]. Pedro et al. also found a twofold increase in phosphorylated Tau protein levels in the hippocampus and frontal cortex of APP/PS1 mice compared to wild-type controls [88]. Recent clinical research indicates that abnormal phosphorylation of Tau protein in the brain occurs before Aβ deposition [89]. Excessive phosphorylation of Tau can induce endoplasmic reticulum stress, synaptic dysfunction, and neurodegeneration, exacerbating the progression of AD [90]. The evidence suggests that the role of abnormally hyperphosphorylated Tau protein is significant in the advancement of AD. Therefore, reducing the levels of hyperphosphorylated Tau protein could effectively alleviate AD progression [91,92]. In recent years, the AMPKα subunit has been found to have a role in reducing Tau aggregation. Researchers such as Kai conducted in vivo and in vitro experiments using diabetic and aging mice, as well as SH-SY5Y cells, to investigate the neuroprotective effects of FGF21 and found that FGF21 can promote AMPKα phosphorylation to inhibit Tau aggregation [38]. Additionally, Dorlors and colleagues discovered that increased expression of AMPKα can reduce phosphorylated Tau aggregation by enhancing autophagy [93]. Apart from contributing to the inhibition of phosphorylated Tau aggregation, the AMPKα subunit also plays a role in inhibiting Tau phosphorylation. Studies have shown that AMPKα deactivates phosphorylation at the Thr172 site of protein phosphatase 2A (PP2A), thereby reducing phosphorylation at the Tau-Ser262 site [39]. In summary, the AMPKα subunit plays a role in regulating Tau protein aggregation and phosphorylation processes. However, the underlying mechanisms are not yet clear, which limits a deeper exploration of the relationship between AMPKα and Tau pathology. Therefore, further exploration of AMPKα-related pathways or target effects on Tau pathology is necessary to gain a clearer understanding of the relationship between the AMPKα subunit and Tau pathology.
3.3. AMPKα subunit and metabolic disorders
It is known that abnormalities in energy metabolism and dysfunction in the hypothalamus are associated with AD [94]. Specifically, hypothalamic dysfunction can impact the progression of AD and contribute to its onset [95]. Mitochondrial dysfunction and endoplasmic reticulum stress induced by AD, in turn, exacerbate the burden on the hypothalamus, further damaging metabolism [90]. Metabolic disturbances link AD with diseases such as diabetes, revealing connections between these conditions. Notably, 80 % of AD patients exhibit impaired glucose tolerance or have type 2 diabetes [96]. Impaired glucose tolerance and insulin resistance have been observed in AD mouse models [97,98]. The risk of dementia in individuals with type 2 diabetes is increased by 1.5–2 times compared to the general population [99]. Insulin resistance in type 2 diabetes also contributes to the deposition of Aβ plaques [100]. The AMPKα subunit regulates energy metabolism in the body, but its role in regulating energy metabolism in AD is not yet fully understood. In recent years, researchers such as Camila have discovered that icariin can restore metabolic disturbances in APP/PS1 mice by restoring AMPKα subunit activity [40], elucidating the role of the AMPKα subunit in metabolic disruptions in AD. Furthermore, other studies have explored the correlation between the AMPKα subunit and insulin resistance. As mentioned earlier, insulin resistance is also present in AD and is associated with the deposition of Aβ plaques. Therefore, targeting insulin resistance for treatment or even prevention of AD may be effective. It is noteworthy that the AMPKα subunit has been shown to be associated with insulin resistance, and its activation in the body can improve insulin resistance and the expression levels of the Glucose Transporter 4 (GLUT4) gene [101]. Additionally, inhibition of the AMPKα-related pathway SIRT1/AMPKα/Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-Alpha (PGC1-α) can lead to insulin resistance and affect energy metabolism [102,103]. The regulation of the AMPKα/Mechanistic Target of Rapamycin (mTOR) pathway is also related to insulin resistance [104,105]. In conclusion, metabolic disturbances are a key aspect of the pathogenesis of AD, serving as a bridge between AD and metabolic diseases such as diabetes. The AMPKα subunit can regulate metabolic disruptions in AD and may hinder the progression of AD pathogenesis by modulating insulin resistance in AD.
3.4. AMPKα subunit and inflammation
Inflammation is one of the characteristics of AD and can lead to neuronal degeneration and loss, fundamentally impacting disease progression [106,107]. AD mouse models exhibit high levels of inflammatory cytokines such as Tumor Necrosis Factor-alpha (TNF-α), Interleukin-1 beta (IL-1β), and Interferon-gamma (INF-γ) [86]. The activation of inflammatory signals can promote the expression of APP, increase the activity of γ-secretase, and result in the release of a large amount of Aβ peptides [108]. The excessive activation of inflammatory responses in AD is intricately linked to the activation of neuroglial cells. Both AD patients and AD mouse models show activation of astrocytes and microglia in the brain regions with Aβ plaques [109]. Caveolin-1 is expressed in various neural cells, including astrocytes, and is involved in the pathogenesis of AD [110]. Researchers such as Gang have found an interaction between AMPKα subunit and Caveolin-1, which induces neuroinflammation. Metformin can modulate this interaction and alleviate the inflammatory response [41], and it is also believed to play a role in alleviating AD-related inflammation [111]. Therefore, the AMPKα subunit may be a key regulatory factor in the neuroinflammatory response associated with astrocytes in the Aβ plaque regions. Furthermore, the AMPKα subunit is associated with the activation of microglia in AD. Inhibiting the release of pro-inflammatory factors in microglia can have anti-inflammatory effects, a process regulated by the AMPKα subunit [112]. In summary, the AMPKα subunit has the ability to regulate the activation of neuroglial cells and inflammatory responses in AD, and it may be a key regulatory factor in the neuroinflammatory response associated with astrocytes in the Aβ plaque regions.
3.5. AMPKα subunit and mitochondrial dysfunction
Mitochondria are important double-membrane organelles in the cytoplasm of eukaryotic cells [113,114], crucial for the production of Adenosine Triphosphate (ATP) and the maintenance of calcium homeostasis [115,116]. The brain is one of the most metabolically active organs in the human body, primarily using glucose as its main energy source. As a high-energy-consuming organ, approximately 93 % of its energy requirements are provided by ATP generated by mitochondria [117]. Oxidative phosphorylation on the inner mitochondrial membrane is the main process for ATP production [118]. Previous studies have found that ATP levels in the brains of AD mice are lower than in wild-type mice, indicating that mitochondrial dysfunction is an important pathogenic mechanism in AD [119]. This viewpoint is receiving increasing attention and prompting exploration of new therapeutic strategies aimed at improving mitochondrial function to enhance AD treatment [[120], [121], [122]]. As mentioned earlier, the AMPKα subunit is the main catalytic subunit of the AMPK complex and plays a crucial role in the activation of the AMPK complex. Recent studies have found a close association between the AMPKα subunit and mitochondrial dysfunction in AD. Mitochondrial proteomic analysis of brain tissues from 5 × FAD mice revealed that changes in the phosphorylation level of AMPKα can increase mitochondrial biogenesis by modulating the AMPK/PGC-1α pathway [42]. Knocking down the AMPKα subunit can block the protective effect of drugs on mitochondrial damage in neurons of APP/PS1 mice, indicating the important role of the AMPKα subunit in AD-related mitochondrial dysfunction [123]. However, research on the association between the AMPKα subunit and mitochondrial dysfunction in AD lacks investigation in other AD models related to Tau, leading to a partial and limited understanding. Overall, current research suggests that the AMPKα subunit is associated with mitochondrial dysfunction in AD and regulates mitochondrial biogenesis through the AMPK/PGC-1α pathway.
3.6. AMPKα subunit and inflammasome
In the central nervous system, the main inflammasomes are nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain-containing protein 1 (NLRP1), nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain-containing protein 3 (NLRP3), and absent in melanoma 2 (AIM2) [124], with NLRP1 and NLRP3 showing high expression in the brain tissues of AD patients [125]. These inflammasomes are regulated by Aβ: on one hand, Aβ can induce neuronal pyroptosis through the NLRP3-caspase-1 signaling pathway by cleaving gasdermin D (GSDMD) [126]; on the other hand, Aβ can increase the levels of primary NLRP1 in cortical neurons, thereby activating caspase-1 signaling [127], indicating that NLRP1 and NLRP3 are key factors in Aβ-mediated neurotoxic effects and play crucial roles in the pathogenesis of AD. Research has shown that knocking down the AMPKα subunit using siRNA can enhance the expression of NLRP3, Apoptosis-associated speck-like protein containing a CARD (ASC), and caspase-1 in SH-SY5Y cells, indicating the role of the AMPKα subunit in inhibiting cell inflammasomes [43]. However, it is currently unknown whether the dysregulation of the AMPKα subunit affects NLRP1 inflammasomes. Furthermore, activation of NLRP3 inflammasomes in microglial cells is associated with AD [128]. Studies have found that activation of AMPKα subunit can suppress activation of spinal cord microglia [129,130] and improve the production of pro-inflammatory mediators by microglia [[131], [132], [133]]. However, whether the effect of AMPKα subunit on microglia is mediated in whole or in part through NLRP3 inflammasomes remains unclear. Overall, there is limited research on the association between the AMPKα subunit and pyroptosis in AD. Given the increasing attention to pyroptosis in the pathogenesis of AD, further research will clarify the relationship between the AMPKα subunit and pyroptosis, potentially identifying meaningful therapeutic targets.
3.7. AMPKα subunit and pyroptosis
Pyroptosis is one of the programmed cell death pathways and a crucial innate immune response in vivo. Typically, its occurrence activates the immune system to eliminate pathogens; however, excessive activation can exacerbate inflammatory reactions [134]. Research has found that gasdermin family member GSDMD, a direct substrate of caspases, executes pyroptosis by inducing membrane permeabilization after caspase cleavage. Moreover, almost all members of the GSDM family have the ability to form pores in membranes, thereby redefining pyroptosis as a form of programmed cell death mediated by GSDMs [135]. Currently, pyroptosis has been implicated in the pathological mechanisms of AD; the inflammasomes and pro-inflammatory cytokines in the pyroptosis activation pathway are linked to AD pathogenesis. Additionally, AD hallmarks such as Aβ and tau proteins have been shown to participate in pyroptosis, although the mechanisms remain unclear [125]. Studies have identified a role for AMPKα subunit in inhibiting neuronal pyroptosis in the context of AD, potentially through inhibiting the activation of NLRP3 inflammasomes [43]. However, research on the association between AMPKα subunit and pyroptosis in AD is limited. Given the increasing attention to pyroptosis in AD pathogenesis, further research will elucidate the relationship between AMPKα subunit and pyroptosis, potentially identifying meaningful therapeutic targets.
4. The subtype-specific role of AMPKα subunit in AD
The AMPKα1 and AMPKα2 subtypes are widely expressed in neurons [55], although AMPK plays an important role in AD, and the AMPKα subunit is the main catalytic subunit, it has not been elucidated which AMPKα subtype is associated with the disease. Therefore, we separately elucidate the subtype-specific actions of AMPKα1 and AMPKα2 in AD.
4.1. AMPKα1 and AD
Recently, The study found that the AMPKα1 subtype has specific actions in AD. Zimmermann and colleagues [46] found that the expression of the AMPKα1 subtype is continuously elevated in the hippocampus of sporadic and familial AD patients, as well as in the Tg19959 APP transgenic mouse model, whereas AMPKα2 expression is decreased in the hippocampus of sporadic AD patients. This change in AMPKα subtype expression appears to be AD-specific, as no changes in AMPKα subtype were found in Lewy body dementia and frontotemporal dementia. Importantly, reducing the expression of AMPKα1 subtype, but not AMPKα2, in hippocampal and cortical neurons effectively rescued synaptic defects and memory decline in an AD mouse model [46]. Zhou and colleagues found that the specific inhibition of mouse AMPKα1 subtype improved age-related memory impairment by reducing the expression of eukaryotic initiation factor 2α (eIF2α) [44], suggesting that the mechanism by which inhibiting the AMPKα1 subtype improves AD symptoms may be related to the decreased expression of eIF2α. However, Lv [136] and colleagues found that the direct activator of AMPKα1 subtype, DW14006, improved learning and memory impairments in APP/PS1 mice. The mechanism involved stimulating the M2 phenotype transformation of microglia and enhancing autophagy to reduce inflammation through the activation of the AMPKα1/Peroxisome Proliferator-Activated Receptor Gamma (PPARγ)/Cluster of Differentiation 36 (CD36) and AMPKα1/Inhibitor of nuclear factor kappa-B (IκB)/NFκB pathways. The dysregulation and complexity of AMPKα1 subtype in AD suggest that AMPKα1 subtype may, similar to the AMPK complex [137], provide neuronal protection when appropriately activated, while long-term and intense stimulation may be detrimental to neurons. In addition to the central nervous system, Wang and colleagues examined changes in AMPKα subtypes in the plasma of patients diagnosed with mild cognitive impairment (MCI) and AD. They found a decrease in AMPKα1 subtype levels in the plasma of MCI and AD patients, while AMPKα2 levels remained unchanged. Furthermore, the levels of AMPKα1 were correlated with characteristic changes in AD cerebrospinal fluid biomarkers (including decreased Aβ42, increased total Tau protein, and phosphorylated Tau protein) [138]. Whether in the central or peripheral system, the evidence above indicates a close association between the AMPKα1 subtype and the pathogenesis and pathology of AD. Its role in the pathogenesis of AD may be similar to the inflammatory response in the body, where appropriate activation through the AMPKα1/PPARγ/CD36 and AMPKα1/IκB/NFκB pathways enhances autophagic responses for protection. However, excessive activation may increase eIF2α levels, leading to learning and memory impairments. Nonetheless, the relevance of the AMPKα2 subtype to AD cannot be ruled out.
4.2. AMPKα2 and AD
Current research indicates a close association between the AMPKα2 subtype and AD. Studies have found that the AMPKα2 subtype is associated with aging [139] and plays a key role in cognition and long-term synaptic plasticity [63]. Conditional knockout mice lacking the AMPKα2 subtype show impaired learning and memory, reduced hippocampal postsynaptic density formation, abnormal dendritic spine morphology, and related synaptic plasticity defects, while knockout of the AMPKα1 subtype has no effect [63]. The role of the AMPKα2 subtype in aging and cognition suggests that it may also have a specific role in AD. Previously, Won and colleagues first discovered that AMPK can negatively regulate the generation of Aβ, and this effect is related to the AMPKα2 subtype. Increased Aβ production was observed after knocking out the AMPKα2 subtype [140]. However, Wang and colleagues found that the expression of the AMPKα2 subtype is positively correlated with Aβ pathology. The AMPKα2 subtype is overactivated in the hippocampus of non-human primate models of AD, and its dysregulation is associated with increased brain Aβ plaque load, elevated soluble Aβ oligomers, and decreased cerebrospinal fluid Aβ42 levels [47]. Given the limited research on the role of the AMPKα2 subtype in regulating Aβ pathology, more research results are needed to evaluate whether the AMPKα2 subtype positively or negatively regulates Aβ pathology. Future studies should further elucidate the specific mechanisms by which the AMPKα2 subtype regulates Aβ production. In addition to Aβ pathology, several studies suggest that AMPK can regulate AD-related Tau protein pathology [141,142]. However, there is currently no research indicating the relationship between the AMPKα2 subtype and AD-related Tau pathology, which could be a potential focus for future research. Furthermore, it is known that diabetes is often associated with cognitive impairment, and severe cognitive impairment can progress to AD [143]. AMPK is considered a bridge between AD and diabetes [144]. Recent research by Li and colleagues found that the activity of the AMPKα2 subtype is inhibited in the hippocampus of mice with diabetes-related brain damage, indicating the involvement of the AMPKα2 subtype in the pathogenesis of diabetes-related brain damage [145]. Therefore, it can be reasonably speculated that the AMPKα2 subtype may be a key factor linking AD and diabetes, but further experiments are needed to verify the accuracy of this speculation. In summary, as the main catalytic subunit of AMPKα in the brain, AMPKα2 plays a crucial role in cognition, learning and memory, and synaptic plasticity. Its ability to regulate Aβ production may make it a potential therapeutic target for AD-related treatments (Fig. 1).
Fig. 1.
Simplified mechanistic representation of the role of AMPKα subunit in the pathogenesis of Alzheimer's disease.
5. Summary and outlook
Despite the fact that AMPK typically has a positive impact on the health of organisms, there is evidence suggesting that it may also be involved in the development of diseases. As emphasized in the introduction, some studies have indicated that AMPK activation has a beneficial effect on AD [28,[32], [33], [34]]. However, several other studies have also reported that AMPK activation exacerbates the progression of AD [4,5,30,31]. Despite the controversy, AMPK is still considered to have potential as a drug target for AD development, possibly due to its significant role in the pathophysiology of AD [[4], [5], [6]] and its potential relevance to the pathogenesis [7]. Multifactorial analysis networks also indicate that AMPK plays a central role in the dysregulation of various metabolic factors and energy homeostasis in AD [146], with dysregulation of its expression leading to downstream changes including amyloid-beta plaque formation, neurofibrillary tangle formation, alterations in metabolic signaling, and memory impairment [146]. Given the existence of different subunits and isoforms of AMPK, comprising a total of 12 AMPK complexes [28], one possible reason for the controversial role of AMPK in AD could be differences in activation among its various subunits or isoforms. Therefore, research is increasingly focusing on exploring the roles of subunits or isoforms within AMPK complexes in AD, as previously mentioned. The AMPKα subunit, as discussed earlier, plays a primary role in AMPK activation [48] and is involved in AD pathology such as Aβ pathology [4,37,38,84,85], Tau pathology [38,39,93], metabolic disturbances [40,[101], [102], [103], [104], [105]], inflammation [41,111,112], mitochondrial dysfunction [42,123], inflammasomes, and pyroptosis [43]. Hence, the AMPKα subunit may represent a potential therapeutic target for AD.
Drug research and development can be viewed as a multi-objective optimization problem [147]. In fact, there have been reports of clinical trials related to AMPKα subunit, such as in type 2 diabetes, where metformin activates AMPKα subunit to upregulate mitochondrial autophagy to prevent further deterioration of blood glucose [148,149]; sorafenib synergistically acts with metformin to activate AMPKα subunit and inhibit non-small cell lung cancer proliferation [150]; Huangqin Qingre Chubi Capsules improve oxidative stress in ankylosing spondylitis patients through the AMPKα/FOXO3a pathway [151]. Additionally, activation of AMPKα subunit is beneficial for the human body; for example, increased exercise activates AMPKα subunit to induce skeletal muscle glucose uptake and protein synthesis [152], and exercise training during fasting activates AMPKα subunit, facilitating rapid activation of muscle protein translation after endurance exercise [153]. However, activation of AMPKα subunit also has certain side effects; for instance, Laura et al. found that chronic activation of AMPK can lead to kidney hypertrophy [154], Dominique et al. discovered increased activation of AMPKα subunit in skeletal muscles of multiple sclerosis patients [155], and Zimmermann et al. found that specific inhibition of AMPKα1 in mouse brains can improve its pathophysiology [46]. The differences in AMPKα subunit observed in different clinical trials may be attributed to variations in disease spectrum or insufficient drug targeting. After all, AMPKα subunit has been found to have subtype-specific effects in AD [45].
It is worth noting that although some studies have noted the subtype-specific effects of the AMPKα subunit in AD, research on the role of AMPKα subtypes in the pathogenesis of AD remains limited. To provide more precise therapeutic targets for AD treatment, further clarification of the role of AMPKα subtypes in AD, including their impact on AD-related pathology and the underlying mechanisms, is needed. Additionally, current research findings are primarily based on animal experiments, which show variability possibly due to differences in the types of AD model mice used and experimental methods employed by researchers. Lastly, the β subunit, serving as a scaffold subunit in AMPK, has previously been suggested to have an alternative role in regulating AMPK activity [36]. However, there is currently a lack of research on the relationship between the β subunit or its subtypes and the pathogenesis of AD, leaving room for future studies to address this gap.
Funding support
This work was supported by the Funds of National Natural Science Foundation of China (81860710, 82060728), Zunyi Science and Technology Bureau (HZ-2023-09), Guizhou Provincial Science and Technology Department (Thousand Talents Program).
CRediT authorship contribution statement
Lingqiong Xia: Writing – original draft. Jianhua Chen: Writing – review & editing. Juan Huang: Writing – review & editing. Xianmei Lin: Writing – original draft. Jingyu Jiang: Writing – review & editing. Tingting Liu: Writing – review & editing. Nanqu Huang: Writing – review & editing. Yong Luo: Writing – review & editing.
Declaration of competing interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Contributor Information
Nanqu Huang, Email: hnq@zmu.edu.cn.
Yong Luo, Email: luoyong@zmu.edu.cn.
References
- 1.Holtzman D.M., Morris J.C., Goate A.M. Alzheimer's disease: the challenge of the second century. Sci. Transl. Med. 2011;3:77sr1. doi: 10.1126/scitranslmed.3002369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.2023 Alzheimer's disease facts and figures. Alzheimers Dement. 2023;19:1598–1695. doi: 10.1002/alz.13016. [DOI] [PubMed] [Google Scholar]
- 3.Wang W., Zhao F., Ma X., Perry G., Zhu X. Mitochondria dysfunction in the pathogenesis of Alzheimer's disease: recent advances. Mol. Neurodegener. 2020;15:30. doi: 10.1186/s13024-020-00376-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ma T., Chen Y., Vingtdeux V., Zhao H., Viollet B., Marambaud P., Klann E. Inhibition of AMP-activated protein kinase signaling alleviates impairments in hippocampal synaptic plasticity induced by amyloid β. J. Neurosci. 2014;34:12230–12238. doi: 10.1523/JNEUROSCI.1694-14.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Mairet-Coello G., Courchet J., Pieraut S., Courchet V., Maximov A., Polleux F. The CAMKK2-AMPK kinase pathway mediates the synaptotoxic effects of Aβ oligomers through Tau phosphorylation. Neuron. 2013;78:94–108. doi: 10.1016/j.neuron.2013.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Wang X., Zimmermann H.R., Ma T. Therapeutic potential of AMP-activated protein kinase in alzheimer's disease. J. Alzheimers Dis. 2019;68:33–38. doi: 10.3233/JAD-181043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Cai Z., Yan L.-J., Li K., Quazi S.H., Zhao B. Roles of AMP-activated protein kinase in Alzheimer's disease. NeuroMolecular Med. 2012;14:1–14. doi: 10.1007/s12017-012-8173-2. [DOI] [PubMed] [Google Scholar]
- 8.Hardie D.G. AMPK--sensing energy while talking to other signaling pathways. Cell Metabol. 2014;20:939–952. doi: 10.1016/j.cmet.2014.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Xiao B., Heath R., Saiu P., Leiper F.C., Leone P., Jing C., Walker P.A., Haire L., Eccleston J.F., Davis C.T., Martin S.R., Carling D., Gamblin S.J. Structural basis for AMP binding to mammalian AMP-activated protein kinase. Nature. 2007;449:496–500. doi: 10.1038/nature06161. [DOI] [PubMed] [Google Scholar]
- 10.Davies S.P., Helps N.R., Cohen P.T., Hardie D.G. 5’-AMP inhibits dephosphorylation, as well as promoting phosphorylation, of the AMP-activated protein kinase. Studies using bacterially expressed human protein phosphatase-2C alpha and native bovine protein phosphatase-2AC. FEBS Lett. 1995;377:421–425. doi: 10.1016/0014-5793(95)01368-7. [DOI] [PubMed] [Google Scholar]
- 11.Molina E., Hong L. Ii. Chefetz, AMPKα-like proteins as LKB1 downstream targets in cell physiology and cancer. J. Mol. Med. (Berl.) 2021;99:651–662. doi: 10.1007/s00109-021-02040-y. [DOI] [PubMed] [Google Scholar]
- 12.Najar M.A., Rex D.a.B., Modi P.K., Agarwal N., Dagamajalu S., Karthikkeyan G., Vijayakumar M., Chatterjee A., Sankar U., Prasad T.S.K. A complete map of the Calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) signaling pathway. J. Cell Commun. Signal. 2021;15:283–290. doi: 10.1007/s12079-020-00592-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Neumann D. Is TAK1 a direct upstream kinase of AMPK? Int. J. Mol. Sci. 2018;19:2412. doi: 10.3390/ijms19082412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zhang C.-S., Hawley S.A., Zong Y., Li M., Wang Z., Gray A., Ma T., Cui J., Feng J.-W., Zhu M., Wu Y.-Q., Li T.Y., Ye Z., Lin S.-Y., Yin H., Piao H.-L., Hardie D.G., Lin S.-C. Fructose-1,6-bisphosphate and aldolase mediate glucose sensing by AMPK. Nature. 2017;548:112–116. doi: 10.1038/nature23275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Salt I.P., Johnson G., Ashcroft S.J., Hardie D.G. AMP-activated protein kinase is activated by low glucose in cell lines derived from pancreatic beta cells, and may regulate insulin release. Biochem. J. 1998;335(Pt 3):533–539. doi: 10.1042/bj3350533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Zhang C.-S., Jiang B., Li M., Zhu M., Peng Y., Zhang Y.-L., Wu Y.-Q., Li T.Y., Liang Y., Lu Z., Lian G., Liu Q., Guo H., Yin Z., Ye Z., Han J., Wu J.-W., Yin H., Lin S.-Y., Lin S.-C. The lysosomal v-ATPase-Ragulator complex is a common activator for AMPK and mTORC1, acting as a switch between catabolism and anabolism. Cell Metabol. 2014;20:526–540. doi: 10.1016/j.cmet.2014.06.014. [DOI] [PubMed] [Google Scholar]
- 17.Pinkosky S.L., Scott J.W., Desjardins E.M., Smith B.K., Day E.A., Ford R.J., Langendorf C.G., Ling N.X.Y., Nero T.L., Loh K., Galic S., Hoque A., Smiles W.J., Ngoei K.R.W., Parker M.W., Yan Y., Melcher K., Kemp B.E., Oakhill J.S., Steinberg G.R. Long-chain fatty acyl-CoA esters regulate metabolism via allosteric control of AMPK β1 isoforms. Nat. Metab. 2020;2:873–881. doi: 10.1038/s42255-020-0245-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Hardie D.G. AMP-activated protein kinase: a cellular energy sensor with a key role in metabolic disorders and in cancer. Biochem. Soc. Trans. 2011;39:1–13. doi: 10.1042/BST0390001. [DOI] [PubMed] [Google Scholar]
- 19.Polekhina G., Gupta A., Michell B.J., van Denderen B., Murthy S., Feil S.C., Jennings I.G., Campbell D.J., Witters L.A., Parker M.W., Kemp B.E., Stapleton D. AMPK beta 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]
- 20.Iseli T.J., Walter M., van Denderen B.J.W., Katsis F., Witters L.A., Kemp B.E., Michell B.J., Stapleton D. AMP-activated protein kinase beta subunit tethers alpha and gamma subunits via its C-terminal sequence (186-270) J. Biol. Chem. 2005;280:13395–13400. doi: 10.1074/jbc.M412993200. [DOI] [PubMed] [Google Scholar]
- 21.McBride A., Ghilagaber S., Nikolaev A., Hardie D.G. The glycogen-binding domain on the AMPK beta subunit allows the kinase to act as a glycogen sensor. Cell Metabol. 2009;9:23–34. doi: 10.1016/j.cmet.2008.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Fu X., Wan S., Lyu Y.L., Liu L.F., Qi H. Etoposide induces ATM-dependent mitochondrial biogenesis through AMPK activation. PLoS One. 2008;3 doi: 10.1371/journal.pone.0002009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Li S., Lavagnino Z., Lemacon D., Kong L., Ustione A., Ng X., Zhang Y., Wang Y., Zheng B., Piwnica-Worms H., Vindigni A., Piston D.W., You Z. Ca2+-Stimulated AMPK-dependent phosphorylation of Exo1 protects stressed replication forks from aberrant resection. Mol. Cell. 2019;74:1123–1137.e6. doi: 10.1016/j.molcel.2019.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Sanli T., Rashid A., Liu C., Harding S., Bristow R.G., Cutz J.-C., Singh G., Wright J., Tsakiridis T. Ionizing radiation activates AMP-activated kinase (AMPK): a target for radiosensitization of human cancer cells. Int. J. Radiat. Oncol. Biol. Phys. 2010;78:221–229. doi: 10.1016/j.ijrobp.2010.03.005. [DOI] [PubMed] [Google Scholar]
- 25.Steinberg G.R., Hardie D.G. New insights into activation and function of the AMPK. Nat. Rev. Mol. Cell Biol. 2023;24:255–272. doi: 10.1038/s41580-022-00547-x. [DOI] [PubMed] [Google Scholar]
- 26.Wu A.Y.-T., Sekar P., Huang D.-Y., Hsu S.-H., Chan C.-M., Lin W.-W. Spatiotemporal roles of AMPK in PARP-1- and autophagy-dependent retinal pigment epithelial cell death caused by UVA. J. Biomed. Sci. 2023;30:91. doi: 10.1186/s12929-023-00978-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Puustinen P., Keldsbo A., Corcelle-Termeau E., Ngoei K., Sønder S.L., Farkas T., Kaae Andersen K., Oakhill J.S., Jäättelä M. DNA-dependent protein kinase regulates lysosomal AMP-dependent protein kinase activation and autophagy. Autophagy. 2020;16:1871–1888. doi: 10.1080/15548627.2019.1710430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.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]
- 29.Huang J., Huang N., Mao Q., Shi J., Qiu Y. Natural bioactive compounds in Alzheimer's disease: from the perspective of type 3 diabetes mellitus. Front. Aging Neurosci. 2023;15 doi: 10.3389/fnagi.2023.1130253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Yoon S.O., Park D.J., Ryu J.C., Ozer H.G., Tep C., Shin Y.J., Lim T.H., Pastorino L., Kunwar A.J., Walton J.C., Nagahara A.H., Lu K.P., Nelson R.J., Tuszynski M.H., Huang K. JNK3 perpetuates metabolic stress induced by Aβ peptides. Neuron. 2012;75:824–837. doi: 10.1016/j.neuron.2012.06.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Chen Y., Zhou K., Wang R., Liu Y., Kwak Y.-D., Ma T., Thompson R.C., Zhao Y., Smith L., Gasparini L., Luo Z., Xu H., Liao F.-F. Antidiabetic drug metformin (GlucophageR) increases biogenesis of Alzheimer's amyloid peptides via up-regulating BACE1 transcription. Proc. Natl. Acad. Sci. U. S. A. 2009;106:3907–3912. doi: 10.1073/pnas.0807991106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Zhou X.-G., Qiu W.-Q., Yu L., Pan R., Teng J.-F., Sang Z.-P., Law B.Y.-K., Zhao Y., Zhang L., Yan L., Tang Y., Sun X.-L., Wong V.K.W., Yu C.-L., Wu J.-M., Qin D.-L., Wu A.-G. Targeting microglial autophagic degradation of the NLRP3 inflammasome for identification of thonningianin A in Alzheimer's disease. Inflamm. Regen. 2022;42:25. doi: 10.1186/s41232-022-00209-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ou Z., Kong X., Sun X., He X., Zhang L., Gong Z., Huang J., Xu B., Long D., Li J., Li Q., Xu L., Xuan A. Metformin treatment prevents amyloid plaque deposition and memory impairment in APP/PS1 mice. Brain Behav. Immun. 2018;69:351–363. doi: 10.1016/j.bbi.2017.12.009. [DOI] [PubMed] [Google Scholar]
- 34.Chiang M.-C., Cheng Y.-C., Chen S.-J., Yen C.-H., Huang R.-N. Metformin activation of AMPK-dependent pathways is neuroprotective in human neural stem cells against Amyloid-beta-induced mitochondrial dysfunction. Exp. Cell Res. 2016;347:322–331. doi: 10.1016/j.yexcr.2016.08.013. [DOI] [PubMed] [Google Scholar]
- 35.Rajamohan F., Reyes A.R., Frisbie R.K., Hoth L.R., Sahasrabudhe P., Magyar R., Landro J.A., Withka J.M., Caspers N.L., Calabrese M.F., Ward J., Kurumbail R.G. Probing the enzyme kinetics, allosteric modulation and activation of α1- and α2-subunit-containing AMP-activated protein kinase (AMPK) heterotrimeric complexes by pharmacological and physiological activators. Biochem. J. 2016;473:581–592. doi: 10.1042/BJ20151051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Sanz P., Rubio T., Garcia-Gimeno M.A. AMPKβ subunits: more than just a scaffold in the formation of AMPK complex. FEBS J. 2013;280:3723–3733. doi: 10.1111/febs.12364. [DOI] [PubMed] [Google Scholar]
- 37.Cai Z.-Y., Wang C.-L., Lu T.-T., Yang W.-M. Berberine alleviates amyloid-beta pathogenesis via activating LKB1/AMPK signaling in the brain of APP/PS1 transgenic mice. Curr. Mol. Med. 2019;19:342–348. doi: 10.2174/1566524019666190315164120. [DOI] [PubMed] [Google Scholar]
- 38.Kang K., Xu P., Wang M., Chunyu J., Sun X., Ren G., Xiao W., Li D. FGF21 attenuates neurodegeneration through modulating neuroinflammation and oxidant-stress. Biomed. Pharmacother. 2020;129 doi: 10.1016/j.biopha.2020.110439. [DOI] [PubMed] [Google Scholar]
- 39.St-Cyr Giguère F., Attiori Essis S., Chagniel L., Germain M., Cyr M., Massicotte G. The sphingosine-1-phosphate receptor 1 agonist SEW2871 reduces Tau-Ser262 phosphorylation in rat hippocampal slices. Brain Res. 2017;1658:51–59. doi: 10.1016/j.brainres.2017.01.014. [DOI] [PubMed] [Google Scholar]
- 40.Gherardelli C., Cisternas P., Vera-Salazar R.F., Mendez-Orellana C., Inestrosa N.C. Age- and sex-associated glucose metabolism decline in a mouse model of alzheimer's disease. J. Alzheimers Dis. 2022;87:901–917. doi: 10.3233/JAD-215273. [DOI] [PubMed] [Google Scholar]
- 41.Wang G., Cui W., Chen S., Shao Z., Li Y., Wang W., Mao L., Li J., Mei X. Metformin alleviates high glucose-induced ER stress and inflammation by inhibiting the interaction between caveolin1 and AMPKα in rat astrocytes. Biochem. Biophys. Res. Commun. 2021;534:908–913. doi: 10.1016/j.bbrc.2020.10.075. [DOI] [PubMed] [Google Scholar]
- 42.Li T., Li X., Huang X., Yu H., Li S., Zhang Z., Xie Y., Song X., Liu J., Yang X., Liu G. Mitochondriomics reveals the underlying neuroprotective mechanism of TrkB receptor agonist R13 in the 5×FAD mice. Neuropharmacology. 2022;204 doi: 10.1016/j.neuropharm.2021.108899. [DOI] [PubMed] [Google Scholar]
- 43.Zhao N., Sun C., Zheng M., Liu S., Shi R. Amentoflavone suppresses amyloid β1-42 neurotoxicity in Alzheimer's disease through the inhibition of pyroptosis. Life Sci. 2019;239 doi: 10.1016/j.lfs.2019.117043. [DOI] [PubMed] [Google Scholar]
- 44.Zhou X., Yang W., Wang X., Ma T. Isoform-specific effects of neuronal repression of the AMPK catalytic subunit on cognitive function in aged mice. Aging (Albany NY) 2023;15:932–946. doi: 10.18632/aging.204554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zhao F., Wang C., Zhu X. Isoform-specific roles of AMPK catalytic α subunits in Alzheimer's disease. J. Clin. Invest. 2020;130:3403–3405. doi: 10.1172/JCI137908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zimmermann H.R., Yang W., Kasica N.P., Zhou X., Wang X., Beckelman B.C., Lee J., Furdui C.M., Keene C.D., Ma T. Brain-specific repression of AMPKα1 alleviates pathophysiology in Alzheimer's model mice. J. Clin. Invest. 2020;130:3511–3527. doi: 10.1172/JCI133982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wang X., Zhou X., Uberseder B., Lee J., Latimer C.S., Furdui C.M., Keene C.D., Montine T.J., Register T.C., Craft S., Shively C.A., Ma T. Isoform-specific dysregulation of AMP-activated protein kinase signaling in a non-human primate model of Alzheimer's disease. Neurobiol. Dis. 2021;158 doi: 10.1016/j.nbd.2021.105463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.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]
- 49.Cantó C., Auwerx J. AMP-activated protein kinase and its downstream transcriptional pathways. Cell. Mol. Life Sci. 2010;67:3407–3423. doi: 10.1007/s00018-010-0454-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Hawley S.A., Davison M., Woods A., Davies S.P., Beri R.K., Carling D., Hardie D.G. 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]
- 51.Stein S.C., Woods A., Jones N.A., Davison M.D., Carling D. The regulation of AMP-activated protein kinase by phosphorylation. Biochem. J. 345 Pt. 2000;3:437–443. doi: 10.1042/bj3450437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Burkewitz K., Zhang Y., Mair W.B. AMPK at the nexus of energetics and aging. Cell Metabol. 2014;20:10–25. doi: 10.1016/j.cmet.2014.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Xiao B., Sanders M.J., Underwood E., Heath R., Mayer F.V., Carmena D., Jing C., Walker P.A., Eccleston J.F., Haire L.F., Saiu P., Howell S.A., Aasland R., Martin S.R., Carling D., Gamblin S.J. 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]
- 54.Steinberg G.R., Kemp B.E. AMPK in health and disease. Physiol. Rev. 2009;89:1025–1078. doi: 10.1152/physrev.00011.2008. [DOI] [PubMed] [Google Scholar]
- 55.Culmsee C., Monnig J., Kemp B.E., Mattson M.P. 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]
- 56.Michell B.J., Stapleton D., Mitchelhill K.I., House C.M., Katsis F., Witters L.A., Kemp B.E. Isoform-specific purification and substrate specificity of the 5’-AMP-activated protein kinase. J. Biol. Chem. 1996;271:28445–28450. doi: 10.1074/jbc.271.45.28445. [DOI] [PubMed] [Google Scholar]
- 57.Jørgensen S.B., Viollet B., Andreelli F., Frøsig C., Birk J.B., Schjerling P., Vaulont S., Richter E.A., Wojtaszewski J.F.P. Knockout of the alpha2 but not alpha1 5’-AMP-activated protein kinase isoform abolishes 5-aminoimidazole-4-carboxamide-1-beta-4-ribofuranosidebut not contraction-induced glucose uptake in skeletal muscle. J. Biol. Chem. 2004;279:1070–1079. doi: 10.1074/jbc.M306205200. [DOI] [PubMed] [Google Scholar]
- 58.Zaha V.G., Young L.H. AMP-activated protein kinase regulation and biological actions in the heart. Circ. Res. 2012;111:800–814. doi: 10.1161/CIRCRESAHA.111.255505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Turnley A.M., Stapleton D., Mann R.J., Witters L.A., Kemp B.E., Bartlett P.F. 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]
- 60.Sun B., He S., Liu B., Xu G., null Guoji E., Feng L., Xu L., Chen D., Zhao W., Chen J., Gao Y., Zhang E. Stanniocalcin-1 protected astrocytes from hypoxic damage through the AMPK pathway. Neurochem. Res. 2021;46:2948–2957. doi: 10.1007/s11064-021-03393-z. [DOI] [PubMed] [Google Scholar]
- 61.Mangalam A.K., Rattan R., Suhail H., Singh J., Hoda M.N., Deshpande M., Fulzele S., Denic A., Shridhar V., Kumar A., Viollet B., Rodriguez M., Giri S. AMP-activated protein kinase suppresses autoimmune central nervous system disease by regulating M1-type macrophage-Th17 Axis. J. Immunol. 2016;197:747–760. doi: 10.4049/jimmunol.1501549. [DOI] [PubMed] [Google Scholar]
- 62.Lin S.-P., Wei J.-X., Hu J.-S., Bu J.-Y., Zhu L.-D., Li Q., Liao H.-J., Lin P.-Y., Ye S., Chen S.-Q., Chen X.-H. Artemisinin improves neurocognitive deficits associated with sepsis by activating the AMPK axis in microglia. Acta Pharmacol. Sin. 2021;42:1069–1079. doi: 10.1038/s41401-021-00634-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Yang W., Zhou X., Zimmermann H.R., Ma T. Brain-specific suppression of AMPKα2 isoform impairs cognition and hippocampal LTP by PERK-mediated eIF2α phosphorylation. Mol. Psychiatr. 2021;26:1880–1897. doi: 10.1038/s41380-020-0739-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Minokoshi Y., Alquier T., Furukawa N., Kim Y.-B., Lee A., Xue B., Mu J., Foufelle F., Ferré P., Birnbaum M.J., Stuck B.J., Kahn B.B. AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature. 2004;428:569–574. doi: 10.1038/nature02440. [DOI] [PubMed] [Google Scholar]
- 65.Viollet B., Andreelli F., Jørgensen S.B., Perrin C., Geloen A., Flamez D., Mu J., Lenzner C., Baud O., Bennoun M., Gomas E., Nicolas G., Wojtaszewski J.F.P., Kahn A., Carling D., Schuit F.C., Birnbaum M.J., Richter E.A., Burcelin R., Vaulont S. The AMP-activated protein kinase alpha2 catalytic subunit controls whole-body insulin sensitivity. J. Clin. Invest. 2003;111:91–98. doi: 10.1172/JCI16567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Xu Z., Liu Q., Li J., Wang J., Yang Z., Wang J., Gao L., Cheng J., He J., Dong Y., Guo X., Cui J., Zhang W. AMPKα is active in autophagy of endothelial cells in arsenic-induced vascular endothelial dysfunction by regulating mTORC1/p70S6K/ULK1. Chem. Biol. Interact. 2024;388 doi: 10.1016/j.cbi.2023.110832. [DOI] [PubMed] [Google Scholar]
- 67.Zhao Y., Lin S., Zeng W., Lin X., Qin X., Miu B., Gao S., Wu H., Liu J., Chen X. JS-K activates G2/M checkpoints through the DNA damage response and induces autophagy via CAMKKβ/AMPKα/mTOR pathway in bladder cancer cells. J. Cancer. 2024;15:343–355. doi: 10.7150/jca.86393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Jin N., Zhang M., Zhou L., Jin S., Cheng H., Li X., Shi Y., Xiang T., Zhang Z., Liu Z., Zhao H., Xie J. Mitochondria transplantation alleviates cardiomyocytes apoptosis through inhibiting AMPKα-mTOR mediated excessive autophagy. Faseb. J. 2024;38 doi: 10.1096/fj.202400375R. [DOI] [PubMed] [Google Scholar]
- 69.Zhang J., Liu Z., Yin X., Wang E., Wang J. NSC48160 targets AMPKα to ameliorate nonalcoholic steatohepatitis by inhibiting lipogenesis and mitochondrial oxidative stress. iScience. 2024;27 doi: 10.1016/j.isci.2023.108614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Hong J., Mei C., Abbas Raza S.H., Khan R., Cheng G., Zan L. SIRT6 cooperates with SIRT5 to regulate bovine preadipocyte differentiation and lipid metabolism via the AMPKα signaling pathway. Arch. Biochem. Biophys. 2020;681 doi: 10.1016/j.abb.2020.108260. [DOI] [PubMed] [Google Scholar]
- 71.H J., N Z., C J., X M., K Q., Y Z., J W. Metabolic and transcriptome responses of RNAi-mediated AMPKα knockdown in Tribolium castaneum. BMC Genom. 2020;21 doi: 10.1186/s12864-020-07070-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Yang W., Zhou X., Wang X., Lee J., Wu D., Sun P., Furdui C.M., Ma T. Protein expression alteration in hippocampus upon genetic repression of AMPKα isoforms. Hippocampus. 2021;31:353–361. doi: 10.1002/hipo.23305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Boland B., Yu W.H., Corti O., Mollereau B., Henriques A., Bezard E., Pastores G.M., Rubinsztein D.C., Nixon R.A., Duchen M.R., Mallucci G.R., Kroemer G., Levine B., Eskelinen E.-L., Mochel F., Spedding M., Louis C., Martin O.R., Millan M.J. Promoting the clearance of neurotoxic proteins in neurodegenerative disorders of ageing. Nat. Rev. Drug Discov. 2018;17:660–688. doi: 10.1038/nrd.2018.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Hur J.-Y. γ-Secretase in Alzheimer's disease. Exp. Mol. Med. 2022;54:433–446. doi: 10.1038/s12276-022-00754-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Sehar U., Rawat P., Reddy A.P., Kopel J., Reddy P.H. Amyloid beta in aging and alzheimer's disease. Int. J. Mol. Sci. 2022;23 doi: 10.3390/ijms232112924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Mitsuishi Y., Nakano M., Kojima H., Okabe T., Nishimura M. Reduction of amyloid-β production without inhibiting secretase activity by MS-275. ACS Chem. Neurosci. 2024;15:1234–1241. doi: 10.1021/acschemneuro.3c00848. [DOI] [PubMed] [Google Scholar]
- 77.Liang Y., Kang X., Zhang H., Xu H., Wu X. Knockdown and inhibition of hippocampal GPR17 attenuates lipopolysaccharide-induced cognitive impairment in mice. J. Neuroinflammation. 2023;20:271. doi: 10.1186/s12974-023-02958-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Huang H., Zhu Y., Liao L., Gao S., Tao Y., Fang X., Lian Y., Gao C. The long-term effects of intermittent theta burst stimulation on Alzheimer's disease-type pathologies in APP/PS1 mice. Brain Res. Bull. 2023;202 doi: 10.1016/j.brainresbull.2023.110735. [DOI] [PubMed] [Google Scholar]
- 79.Piccialli I., Greco F., Roviello G., Sisalli M.J., Tedeschi V., di Mola A., Borbone N., Oliviero G., De Feo V., Secondo A., Massa A., Pannaccione A. The 3-(3-oxoisoindolin-1-yl)pentane-2,4-dione (ISOAC1) as a new molecule able to inhibit Amyloid β aggregation and neurotoxicity. Biomed. Pharmacother. 2023;168 doi: 10.1016/j.biopha.2023.115745. [DOI] [PubMed] [Google Scholar]
- 80.Cui Z., Qu L., Zhang Q., Lu F., Liu F. Brazilin-7-2-butenoate inhibits amyloid β-protein aggregation, alleviates cytotoxicity, and protects Caenorhabditis elegans. Int. J. Biol. Macromol. 2024;264 doi: 10.1016/j.ijbiomac.2024.130695. [DOI] [PubMed] [Google Scholar]
- 81.Halipi V., Sasanian N., Feng J., Hu J., Lubart Q., Bernson D., van Leeuwen D., Ahmadpour D., Sparr E., Esbjörner E.K. Extracellular vesicles slow down aβ(1-42) aggregation by interfering with the amyloid fibril elongation step. ACS Chem. Neurosci. 2024;15:944–954. doi: 10.1021/acschemneuro.3c00655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Abutarboush R., Reed E., Chen Y., Gu M., Watson C., Kawoos U., Statz J.K., Tschiffely A.E., Ciarlone S., Perez-Garcia G., Gama Sosa M.A., de Gasperi R., Stone J.R., Elder G.A., Ahlers S.T. Exposure to low-intensity blast increases clearance of brain amyloid beta. J. Neurotrauma. 2024;41:685–704. doi: 10.1089/neu.2023.0284. [DOI] [PubMed] [Google Scholar]
- 83.Zhao Q., Maci M., Miller M.R., Zhou H., Zhang F., Algamal M., Lee Y.F., Hou S.S., Perle S.J., Le H., Russ A.N., Lo E.H., Gerashchenko D., Gomperts S.N., Bacskai B.J., Kastanenka K.V. Sleep restoration by optogenetic targeting of GABAergic neurons reprograms microglia and ameliorates pathological phenotypes in an Alzheimer's disease model. Mol. Neurodegener. 2023;18:93. doi: 10.1186/s13024-023-00682-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Grimm M.O.W., Rothhaar T.L., Hartmann T. The role of APP proteolytic processing in lipid metabolism. Exp. Brain Res. 2012;217:365–375. doi: 10.1007/s00221-011-2975-6. [DOI] [PubMed] [Google Scholar]
- 85.Park S.Y., Lee H.R., Lee W.S., Shin H.K., Kim H.Y., Hong K.W., Kim C.D. Cilostazol modulates autophagic degradation of β-amyloid peptide via SIRT1-coupled LKB1/ampkα signaling in neuronal cells. PLoS One. 2016;11 doi: 10.1371/journal.pone.0160620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Zangerolamo L., Solon C., Soares G.M., Engel D.F., Velloso L.A., Boschero A.C., Carneiro E.M., Barbosa H.C.L. Energy homeostasis deregulation is attenuated by TUDCA treatment in streptozotocin-induced Alzheimer's disease mice model. Sci. Rep. 2021;11 doi: 10.1038/s41598-021-97624-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Radde R., Bolmont T., Kaeser S.A., Coomaraswamy J., Lindau D., Stoltze L., Calhoun M.E., Jäggi F., Wolburg H., Gengler S., Haass C., Ghetti B., Czech C., Hölscher C., Mathews P.M., Jucker M. Abeta42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology. EMBO Rep. 2006;7:940–946. doi: 10.1038/sj.embor.7400784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Dionísio P.A., Amaral J.D., Ribeiro M.F., Lo A.C., D'Hooge R., Rodrigues C.M.P. Amyloid-β pathology is attenuated by tauroursodeoxycholic acid treatment in APP/PS1 mice after disease onset. Neurobiol. Aging. 2015;36:228–240. doi: 10.1016/j.neurobiolaging.2014.08.034. [DOI] [PubMed] [Google Scholar]
- 89.Ye J., Wan H., Chen S., Liu G.-P. Targeting tau in Alzheimer's disease: from mechanisms to clinical therapy. Neural Regen. Res. 2024;19:1489–1498. doi: 10.4103/1673-5374.385847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Ochiai T., Nagayama T., Matsui K., Amano K., Sano T., Wakabayashi T., Iwatsubo T. Tauroursodeoxycholic acid attenuates diet-induced and age-related peripheral endoplasmic reticulum stress and cerebral amyloid pathology in a mouse model of alzheimer's disease. J. Prev. Alzheimers Dis. 2021;8:483–494. doi: 10.14283/jpad.2021.33. [DOI] [PubMed] [Google Scholar]
- 91.Dai C.-L., Tung Y.C., Liu F., Gong C.-X., Iqbal K. Tau passive immunization inhibits not only tau but also Aβ pathology. Alzheimer's Res. Ther. 2017;9:1. doi: 10.1186/s13195-016-0227-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Huang W., Huang J., Huang N., Luo Y. The role of TREM2 in Alzheimer's disease: from the perspective of Tau. Front. Cell Dev. Biol. 2023;11 doi: 10.3389/fcell.2023.1280257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Puigoriol-Illamola D., Griñán-Ferré C., Vasilopoulou F., Leiva R., Vázquez S., Pallàs M. 11β-HSD1 inhibition by RL-118 promotes autophagy and correlates with reduced oxidative stress and inflammation, enhancing cognitive performance in SAMP8 mouse model. Mol. Neurobiol. 2018;55:8904–8915. doi: 10.1007/s12035-018-1026-8. [DOI] [PubMed] [Google Scholar]
- 94.Clarke J.R., Lyra E Silva N.M., Figueiredo C.P., Frozza R.L., Ledo J.H., Beckman D., Katashima C.K., Razolli D., Carvalho B.M., Frazão R., Silveira M.A., Ribeiro F.C., Bomfim T.R., Neves F.S., Klein W.L., Medeiros R., LaFerla F.M., Carvalheira J.B., Saad M.J., Munoz D.P., Velloso L.A., Ferreira S.T., De Felice F.G. Alzheimer-associated Aβ oligomers impact the central nervous system to induce peripheral metabolic deregulation. EMBO Mol. Med. 2015;7:190–210. doi: 10.15252/emmm.201404183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Vercruysse P., Vieau D., Blum D., Petersén Å., Dupuis L. Hypothalamic alterations in neurodegenerative diseases and their relation to abnormal energy metabolism. Front. Mol. Neurosci. 2018;11:2. doi: 10.3389/fnmol.2018.00002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Janson J., Laedtke T., Parisi J.E., O'Brien P., Petersen R.C., Butler P.C. Increased risk of type 2 diabetes in Alzheimer disease. Diabetes. 2004;53:474–481. doi: 10.2337/diabetes.53.2.474. [DOI] [PubMed] [Google Scholar]
- 97.Macklin L., Griffith C.M., Cai Y., Rose G.M., Yan X.-X., Patrylo P.R. Glucose tolerance and insulin sensitivity are impaired in APP/PS1 transgenic mice prior to amyloid plaque pathogenesis and cognitive decline. Exp. Gerontol. 2017;88:9–18. doi: 10.1016/j.exger.2016.12.019. [DOI] [PubMed] [Google Scholar]
- 98.Zangerolamo L., Vettorazzi J.F., Solon C., Bronczek G.A., Engel D.F., Kurauti M.A., Soares G.M., Rodrigues K.S., Velloso L.A., Boschero A.C., Carneiro E.M., Barbosa H.C.L. The bile acid TUDCA improves glucose metabolism in streptozotocin-induced Alzheimer's disease mice model. Mol. Cell. Endocrinol. 2021;521 doi: 10.1016/j.mce.2020.111116. [DOI] [PubMed] [Google Scholar]
- 99.Biessels G.J., Strachan M.W.J., Visseren F.L.J., Kappelle L.J., Whitmer R.A. Dementia and cognitive decline in type 2 diabetes and prediabetic stages: towards targeted interventions. Lancet Diabetes Endocrinol. 2014;2:246–255. doi: 10.1016/S2213-8587(13)70088-3. [DOI] [PubMed] [Google Scholar]
- 100.Beeler N., Riederer B.M., Waeber G., Abderrahmani A. Role of the JNK-interacting protein 1/islet brain 1 in cell degeneration in Alzheimer disease and diabetes. Brain Res. Bull. 2009;80:274–281. doi: 10.1016/j.brainresbull.2009.07.006. [DOI] [PubMed] [Google Scholar]
- 101.Nasrollahi Z., ShahaniPour K., Monajemi R., Ahadi A.M. Effect of quercetin and Abelmoschus esculentus (L.) Moench on lipids metabolism and blood glucose through AMPK-α in diabetic rats (HFD/STZ) J. Food Biochem. 2022;46 doi: 10.1111/jfbc.14506. [DOI] [PubMed] [Google Scholar]
- 102.Fan D., Pan K., Guo J., Liu Z., Zhang C., Zhang J., Qian X., Shen H., Zhao J. Exercise ameliorates fine particulate matter-induced metabolic damage through the SIRT1/AMPKα/PGC1-α/NRF1 signaling pathway. Environ. Res. 2024;245 doi: 10.1016/j.envres.2023.117973. [DOI] [PubMed] [Google Scholar]
- 103.Ye Z., Ma J., Liu Y., Xu B., Dai X., Fu M., Tian T., Sui X., Mo F., Gao S., Zhao D., Zhang D. Jiangtang Sanhao formula ameliorates skeletal muscle insulin resistance via regulating GLUT4 translocation in diabetic mice. Front. Pharmacol. 2022;13 doi: 10.3389/fphar.2022.950535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Gong K., Zhang Z., Chen S.-S., Zhu X.-R., Wang M.-Y., Yang X.-Y., Ding C., Han J.-H., Li Q.-S., Duan Y.-J. 6-Methyl flavone inhibits Nogo-B expression and improves high fructose diet-induced liver injury in mice. Acta Pharmacol. Sin. 2023;44:2216–2229. doi: 10.1038/s41401-023-01121-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Ji A., Chen W., Liu C., Zhang T., Shi R., Wang X., Xu H., Li D. Soy protein compared with whey protein ameliorates insulin resistance by regulating lipid metabolism, AMPK/mTOR pathway and gut microbiota in high-fat diet-fed mice. Food Funct. 2023;14:5752–5767. doi: 10.1039/d3fo01093g. [DOI] [PubMed] [Google Scholar]
- 106.Kinney J.W., Bemiller S.M., Murtishaw A.S., Leisgang A.M., Salazar A.M., Lamb B.T. Inflammation as a central mechanism in Alzheimer's disease. Alzheimers Dement (N Y) 2018;4:575–590. doi: 10.1016/j.trci.2018.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Huang N.-Q., Jin H., Zhou S.-Y., Shi J.-S., Jin F. TLR4 is a link between diabetes and Alzheimer's disease. Behav. Brain Res. 2017;316:234–244. doi: 10.1016/j.bbr.2016.08.047. [DOI] [PubMed] [Google Scholar]
- 108.Decourt B., Lahiri D.K., Sabbagh M.N. Targeting tumor Necrosis factor alpha for alzheimer's disease. Curr. Alzheimer Res. 2017;14:412–425. doi: 10.2174/1567205013666160930110551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Nunes A.F., Amaral J.D., Lo A.C., Fonseca M.B., Viana R.J.S., Callaerts-Vegh Z., D'Hooge R., Rodrigues C.M.P. TUDCA, a bile acid, attenuates amyloid precursor protein processing and amyloid-β deposition in APP/PS1 mice. Mol. Neurobiol. 2012;45:440–454. doi: 10.1007/s12035-012-8256-y. [DOI] [PubMed] [Google Scholar]
- 110.Bonds J.A., Shetti A., Bheri A., Chen Z., Disouky A., Tai L., Mao M., Head B.P., Bonini M.G., Haus J.M., Minshall R.D., Lazarov O. Depletion of caveolin-1 in type 2 diabetes model induces alzheimer's disease pathology precursors. J. Neurosci. 2019;39:8576–8583. doi: 10.1523/JNEUROSCI.0730-19.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Ponce-Lopez T., González Álvarez Tostado J.A., Dias F., Montiel Maltez K.H. Metformin prevents NDEA-induced memory impairments associated with attenuating beta-amyloid, tumor Necrosis factor-alpha, and interleukin-6 levels in the Hippocampus of rats. Biomolecules. 2023;13:1289. doi: 10.3390/biom13091289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Wu L.-H., Lin C., Lin H.-Y., Liu Y.-S., Wu C.Y.-J., Tsai C.-F., Chang P.-C., Yeh W.-L., Lu D.-Y. Naringenin suppresses neuroinflammatory responses through inducing suppressor of cytokine signaling 3 expression. Mol. Neurobiol. 2016;53:1080–1091. doi: 10.1007/s12035-014-9042-9. [DOI] [PubMed] [Google Scholar]
- 113.Nunnari J., Suomalainen A. Mitochondria: in sickness and in health. Cell. 2012;148:1145–1159. doi: 10.1016/j.cell.2012.02.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Joshi A., Richard T.H., Gohil V.M. Mitochondrial phospholipid metabolism in health and disease. J. Cell Sci. 2023;136 doi: 10.1242/jcs.260857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Rangaraju V., Lewis T.L., Hirabayashi Y., Bergami M., Motori E., Cartoni R., Kwon S.-K., Courchet J. Pleiotropic mitochondria: the influence of mitochondria on neuronal development and disease. J. Neurosci. 2019;39:8200–8208. doi: 10.1523/JNEUROSCI.1157-19.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Atlante A., Valenti D. Mitochondria have made a long evolutionary path from ancient bacteria immigrants within eukaryotic cells to essential cellular hosts and key players in human health and disease. Curr. Issues Mol. Biol. 2023;45:4451–4479. doi: 10.3390/cimb45050283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Harris J.J., Jolivet R., Attwell D. Synaptic energy use and supply. Neuron. 2012;75:762–777. doi: 10.1016/j.neuron.2012.08.019. [DOI] [PubMed] [Google Scholar]
- 118.Ahmad W., Ijaz B., Shabbiri K., Ahmed F., Rehman S. Oxidative toxicity in diabetes and Alzheimer's disease: mechanisms behind ROS/RNS generation. J. Biomed. Sci. 2017;24:76. doi: 10.1186/s12929-017-0379-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Zhang C., Rissman R.A., Feng J. Characterization of ATP alternations in an Alzheimer's disease transgenic mouse model. J. Alzheimers Dis. 2015;44:375–378. doi: 10.3233/JAD-141890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Wang M., Xuan T., Li H., An J., Hao T., Cheng J. Protective effect of FXN overexpression on ferroptosis in L-Glu-induced SH-SY5Y cells. Acta Histochem. 2024;126 doi: 10.1016/j.acthis.2024.152135. [DOI] [PubMed] [Google Scholar]
- 121.Chen H., Xing H., Zhong C., Lin X., Chen R., Luo N., Chen L., Huang Y. METTL3 confers protection against mitochondrial dysfunction and cognitive impairment in an Alzheimer disease mouse model by upregulating Mfn2 via N6-methyladenosine modification. J. Neuropathol. Exp. Neurol. 2024 doi: 10.1093/jnen/nlae010. [DOI] [PubMed] [Google Scholar]
- 122.Hao L., Shi M., Ma J., Shao S., Yuan Y., Liu J., Yu Z., Zhang Z., Hölscher C., Zhang Z. A cholecystokinin analogue ameliorates cognitive deficits and regulates mitochondrial dynamics via the AMPK/Drp1 pathway in APP/PS1 mice. J. Prev. Alzheimers Dis. 2024;11:382–401. doi: 10.14283/jpad.2024.6. [DOI] [PubMed] [Google Scholar]
- 123.Wang D., Liu X., Liu Y., Li S., Wang C. The effects of cardiotrophin-1 on early synaptic mitochondrial dysfunction and synaptic pathology in APPswe/PS1dE9 mice. J. Alzheimers Dis. 2017;59:1255–1267. doi: 10.3233/JAD-170100. [DOI] [PubMed] [Google Scholar]
- 124.Yap J.K.Y., Pickard B.S., Chan E.W.L., Gan S.Y. The role of neuronal NLRP1 inflammasome in alzheimer's disease: bringing neurons into the neuroinflammation game. Mol. Neurobiol. 2019;56:7741–7753. doi: 10.1007/s12035-019-1638-7. [DOI] [PubMed] [Google Scholar]
- 125.Shen H., Han C., Yang Y., Guo L., Sheng Y., Wang J., Li W., Zhai L., Wang G., Guan Q. Pyroptosis executive protein GSDMD as a biomarker for diagnosis and identification of Alzheimer's disease. Brain Behav. 2021;11 doi: 10.1002/brb3.2063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Han C., Yang Y., Guan Q., Zhang X., Shen H., Sheng Y., Wang J., Zhou X., Li W., Guo L., Jiao Q. New mechanism of nerve injury in Alzheimer's disease: β-amyloid-induced neuronal pyroptosis. J. Cell Mol. Med. 2020;24:8078–8090. doi: 10.1111/jcmm.15439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Tan M.-S., Tan L., Jiang T., Zhu X.-C., Wang H.-F., Jia C.-D., Yu J.-T. Amyloid-β induces NLRP1-dependent neuronal pyroptosis in models of Alzheimer's disease. Cell Death Dis. 2014;5 doi: 10.1038/cddis.2014.348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Terzioglu G., Young-Pearse T.L. Microglial function, INPP5D/SHIP1 signaling, and NLRP3 inflammasome activation: implications for Alzheimer's disease. Mol. Neurodegener. 2023;18:89. doi: 10.1186/s13024-023-00674-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Jiang J., Xu L., Yang L., Liu S., Wang Z. Mitochondrial-derived peptide MOTS-c ameliorates spared nerve injury-induced neuropathic pain in mice by inhibiting microglia activation and neuronal oxidative damage in the spinal cord via the AMPK pathway. ACS Chem. Neurosci. 2023;14:2362–2374. doi: 10.1021/acschemneuro.3c00140. [DOI] [PubMed] [Google Scholar]
- 130.Lin W., Chen W., Liu K., Ma P., Qiu P., Zheng C., Zhang X., Tan P., Xi X., He X. Mitigation of microglia-mediated acute neuroinflammation and tissue damage by heme oxygenase 1 in a rat spinal cord injury model. Neuroscience. 2021;457:27–40. doi: 10.1016/j.neuroscience.2020.08.001. [DOI] [PubMed] [Google Scholar]
- 131.Velagapudi R., Jamshaid F., Lepiarz I., Katola F.O., Hemming K., Olajide O.A. The tiliroside derivative, 3-O-[(E)-(2-oxo-4-(p-tolyl) but-3-en-1-yl] kaempferol produced inhibition of neuroinflammation and activation of AMPK and Nrf2/HO-1 pathways in BV-2 microglia. Int. Immunopharm. 2019;77 doi: 10.1016/j.intimp.2019.105951. [DOI] [PubMed] [Google Scholar]
- 132.Velagapudi R., El-Bakoush A., Lepiarz I., Ogunrinade F., Olajide O.A. AMPK and SIRT1 activation contribute to inhibition of neuroinflammation by thymoquinone in BV2 microglia. Mol. Cell. Biochem. 2017;435:149–162. doi: 10.1007/s11010-017-3064-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Tsai C.-F., Kuo Y.-H., Yeh W.-L., Wu C.Y.-J., Lin H.-Y., Lai S.-W., Liu Y.-S., Wu L.-H., Lu J.-K., Lu D.-Y. Regulatory effects of caffeic acid phenethyl ester on neuroinflammation in microglial cells. Int. J. Mol. Sci. 2015;16:5572–5589. doi: 10.3390/ijms16035572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Man S.M., Karki R., Kanneganti T.-D. Molecular mechanisms and functions of pyroptosis, inflammatory caspases and inflammasomes in infectious diseases. Immunol. Rev. 2017;277:61–75. doi: 10.1111/imr.12534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Shi J., Gao W., Shao F. Pyroptosis: gasdermin-mediated programmed necrotic cell death. Trends Biochem. Sci. 2017;42:245–254. doi: 10.1016/j.tibs.2016.10.004. [DOI] [PubMed] [Google Scholar]
- 136.Lv J., Wang W., Zhu X., Xu X., Yan Q., Lu J., Shi X., Wang Z., Zhou J., Huang X., Wang J., Duan W., Shen X. DW14006 as a direct AMPKα1 activator improves pathology of AD model mice by regulating microglial phagocytosis and neuroinflammation. Brain Behav. Immun. 2020;90:55–69. doi: 10.1016/j.bbi.2020.07.041. [DOI] [PubMed] [Google Scholar]
- 137.Li J., McCullough L.D. Effects of AMP-activated protein kinase in cerebral ischemia. J. Cerebr. Blood Flow Metabol. 2010;30:480–492. doi: 10.1038/jcbfm.2009.255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Wang X., Zimmermann H.R., Lockhart S.N., Craft S., Ma T. Decreased levels of blood AMPKα1 but not AMPKα2 isoform in patients with mild cognitive impairment and alzheimer's disease: a pilot study. J. Alzheimers Dis. 2020;76:217–224. doi: 10.3233/JAD-191189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Wu H., Sun H., He Z., Chen X., Li Y., Zhao X., Kong W., Kong W. The effect and mechanism of 19S proteasome PSMD11/Rpn6 subunit in D-Galactose induced mimetic aging models. Exp. Cell Res. 2020;394 doi: 10.1016/j.yexcr.2020.112093. [DOI] [PubMed] [Google Scholar]
- 140.Won J.-S., Im Y.-B., Kim J., Singh A.K., 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]
- 141.Wang L., Shi F.-X., Li N., Cao Y., Lei Y., Wang J.-Z., Tian Q., Zhou X.-W. AMPK ameliorates tau acetylation and memory impairment through Sirt1. Mol. Neurobiol. 2020;57:5011–5025. doi: 10.1007/s12035-020-02079-x. [DOI] [PubMed] [Google Scholar]
- 142.Wang L., Li N., Shi F.-X., Xu W.-Q., Cao Y., Lei Y., Wang J.-Z., Tian Q., Zhou X.-W. Upregulation of AMPK ameliorates alzheimer's disease-like tau pathology and memory impairment. Mol. Neurobiol. 2020;57:3349–3361. doi: 10.1007/s12035-020-01955-w. [DOI] [PubMed] [Google Scholar]
- 143.Jais A., Solas M., Backes H., Chaurasia B., Kleinridders A., Theurich S., Mauer J., Steculorum S.M., Hampel B., Goldau J., Alber J., Förster C.Y., Eming S.A., Schwaninger M., Ferrara N., Karsenty G., Brüning J.C. Myeloid-cell-derived VEGF maintains brain glucose uptake and limits cognitive impairment in obesity. Cell. 2016;165:882–895. doi: 10.1016/j.cell.2016.03.033. [DOI] [PubMed] [Google Scholar]
- 144.Chen M., Huang N., Liu J., Huang J., Shi J., Jin F. AMPK: a bridge between diabetes mellitus and Alzheimer's disease. Behav. Brain Res. 2021;400 doi: 10.1016/j.bbr.2020.113043. [DOI] [PubMed] [Google Scholar]
- 145.Li Y., Xiang Q., Yao Y.-H., Li J.-J., Wang Y., Li X.-H. Inactivated AMPK-α2 promotes the progression of diabetic brain damage by Cdk5 phosphorylation at Thr485 site. Biochimie. 2020;168:277–284. doi: 10.1016/j.biochi.2019.11.010. [DOI] [PubMed] [Google Scholar]
- 146.Caberlotto L., Lauria M., Nguyen T.-P., Scotti M. The central role of AMP-kinase and energy homeostasis impairment in Alzheimer's disease: a multifactor network analysis. PLoS One. 2013;8 doi: 10.1371/journal.pone.0078919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Huang N., Huang W., Wu J., Long S., Luo Y., Huang J. Possible opportunities and challenges for traditional Chinese medicine research in 2035. Front. Pharmacol. 2024;15 doi: 10.3389/fphar.2024.1426300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Bhansali S., Bhansali A., Dutta P., Walia R., Dhawan V. Metformin upregulates mitophagy in patients with T2DM: a randomized placebo-controlled study. J. Cell Mol. Med. 2020;24:2832–2846. doi: 10.1111/jcmm.14834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Huang J., Huang N., Cui D., Shi J., Qiu Y. Clinical antidiabetic medication used in Alzheimer's disease: from basic discovery to therapeutics development. Front. Aging Neurosci. 2023;15 doi: 10.3389/fnagi.2023.1122300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Groenendijk F.H., Mellema W.W., van der Burg E., Schut E., Hauptmann M., Horlings H.M., Willems S.M., van den Heuvel M.M., Jonkers J., Smit E.F., Bernards R. Sorafenib synergizes with metformin in NSCLC through AMPK pathway activation. Int. J. Cancer. 2015;136:1434–1444. doi: 10.1002/ijc.29113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Huang D., Liu J., Zong R.-K., Wan L. [Huangqin Qingre Chubi Capsules in improving oxidative stress of patients with ankylosing spondylitis via activating PPARγ mediated AMPK/FOXO3a pathway] Zhongguo Zhongyao Zazhi. 2020;45:451–456. doi: 10.19540/j.cnki.cjcmm.20190619.501. [DOI] [PubMed] [Google Scholar]
- 152.Ahtiainen J.P., Walker S., Silvennoinen M., Kyröläinen H., Nindl B.C., Häkkinen K., Nyman K., Selänne H., Hulmi J.J. Exercise type and volume alter signaling pathways regulating skeletal muscle glucose uptake and protein synthesis. Eur. J. Appl. Physiol. 2015;115:1835–1845. doi: 10.1007/s00421-015-3155-3. [DOI] [PubMed] [Google Scholar]
- 153.Van Proeyen K., De Bock K., Hespel P. Training in the fasted state facilitates re-activation of eEF2 activity during recovery from endurance exercise. Eur. J. Appl. Physiol. 2011;111:1297–1305. doi: 10.1007/s00421-010-1753-7. [DOI] [PubMed] [Google Scholar]
- 154.Wilson L., Pollard A.E., Penfold L., Muckett P.J., Whilding C., Bohlooly-Y M., Wilson P., Carling D. Chronic activation of AMP-activated protein kinase leads to early-onset polycystic kidney phenotype. Clin. Sci. (Lond.) 2021;135:2393–2408. doi: 10.1042/CS20210821. [DOI] [PubMed] [Google Scholar]
- 155.Hansen D., Wens I., Vandenabeele F., Verboven K., Eijnde B.O. Altered signaling for mitochondrial and myofibrillar biogenesis in skeletal muscles of patients with multiple sclerosis. Transl. Res. 2015;166:70–79. doi: 10.1016/j.trsl.2015.01.006. [DOI] [PubMed] [Google Scholar]

