Skip to main content

Fueling neurodegeneration: metabolic insights into microglia functions

Abstract

Microglia, the resident immune cells of the central nervous system, emerge in the brain during early embryonic development and persist throughout life. They play essential roles in brain homeostasis, and their dysfunction contributes to neuroinflammation and the progression of neurodegenerative diseases. Recent studies have uncovered an intricate relationship between microglia functions and metabolic processes, offering fresh perspectives on disease mechanisms and possible treatments. Despite these advancements, there are still significant gaps in our understanding of how metabolic dysregulation affects microglial phenotypes in these disorders. This review aims to address these gaps, laying the groundwork for future research on the topic. We specifically examine how metabolic shifts in microglia, such as the transition from oxidative phosphorylation and mitochondrial metabolism to heightened glycolysis during proinflammatory states, impact the disease progression in Alzheimer’s disease, multiple sclerosis, Parkinson’s disease, amyotrophic lateral sclerosis, and Huntington’s disease. Additionally, we explore the role of iron, fatty and amino acid metabolism in microglial homeostasis and repair. Identifying both distinct and shared metabolic adaptations in microglia across neurodegenerative diseases could reveal common therapeutic targets and provide a deeper understanding of disease-specific mechanisms underlying multiple CNS disorders.

Microglia, the resident mononuclear phagocytes of the central nervous system (CNS), are essential components of the brain’s immune system. Derived from erythromyeloid progenitors in the yolk sac, microglia populate the brain and spinal cord during the early embryonic period and constitute 5–15% of the total glial cell population within the CNS parenchyma [1, 2]. These highly specialized cells exhibit unique characteristics during homeostasis, including a slower proliferation rate and a ‘ramified’ morphology, characterized by highly branched processes [3, 4]. During inflammation, microglia rapidly assume ‘hypertrophic morphology’, often referred to as being “activated”, a phenotype characterized by clonal proliferation, an enlarged cell body, and shortened cellular processes [5, 6]. Microglia play two primary roles in the CNS: providing immune defense and maintaining CNS homeostasis. As sentinels, microglia detect signs of pathogenic invasion or tissue damage, playing a crucial role in maintaining the immune integrity of the brain [7]. Simultaneously, they balance the immune response with the ability to moderate potential CNS damage and support tissue repair. In the context of neurodegenerative disorders, however, abnormal activation of microglia exacerbates pathological processes, ultimately resulting in neuronal impairment and neurodegeneration.

Neurodegeneration, which refers to the progressive loss of neural tissues and the function of neurons, is associated with a wide range of symptoms, including cognitive impairment, motor deficits, and dementia [8]. Alzheimer’s disease (AD), multiple sclerosis (MS), Parkinson’s disease (PD), and Huntington’s disease (HD) are among the most prevalent neurodegenerative disorders and affect millions of people, resulting in substantial mortality and morbidity worldwide [9, 10]. While genetic and environmental factors contribute to the etiology of neurodegenerative disorders, age stands out as the most significant risk factor for neurodegeneration. As life expectancy increases, the prevalence of neurodegenerative disorders is expected to rise substantially [11]. Management of such disorders, which are deemed incurable owing to the inability of neurons to self-regenerate after cell death, is limited by a lack of therapies that prevent disease progression.

Chronic neuroinflammation underlying the neurodegeneration is often associated with ‘dysfunctional’ microglia [12,13,14]. As effector cells, microglia are sensitive to pathological stimuli and changes in the microenvironment of the CNS, and their response impacts neuronal networks and contributes to the development of neurodegenerative disorders. As microglia engage in immune responses, they undergo concurrent metabolic changes that closely link these cells to their activation, differentiation, and overall function. This has resulted in growing evidence that highlights the importance of investigating immunometabolism to inform future research and treatments for neurodegenerative diseases [16, 17]. Here, we provide a comprehensive review of the metabolic characteristics of microglia and their importance in driving pathology across different neurodegenerative disorders. By recognizing shared and distinct metabolic adaptations in microglia across neurodegenerative diseases, this review will offer critical insights into how common metabolic dysregulation drives neurodegeneration. These discoveries could pave the way for potentially common therapeutic strategies, advancing both disease-specific and pan-neurodegenerative treatments.

Alzheimer’s disease (AD)

As a widely occurring neurodegenerative disorder, AD manifests in both genetic and sporadic forms. From a biological and pathological perspective, AD is characterized by the accumulation of extracellular plaques composed of amyloid-β (Aβ), followed by the formation of intracellular neurofibrillary tangles made up of tau protein (Fig. 1) [19, 20]. These pathological processes gradually lead to the loss of synapses, the connection between different neurons. The current understanding acknowledges that microglia play a dual role in the development of AD. Initially, microglia are recruited to sites of pathological Aβ plaque deposition to facilitate Aβ clearance and constitute a protective barrier against the neurotoxic effects of Aβ plaques by limiting their outward expansion and contact with nearby Fig. 1.

Fig. 1
figure 1

Key pathological features of neurodegenerative diseases highlight key metabolic events in microglia. This figure illustrates the core metabolic changes in microglia that are common across several neurodegenerative diseases. The figure also highlights disease-specific pathway changes within microglia in each of the listed CNS disorders

healthy neurons [21,22,23]. On the other hand, sustained microglia activation, which results in increased inflammasome activation and the secretion of inflammatory cytokines, has detrimental effects on the surrounding brain tissue, leading to irreversible neurodegeneration [24,25,26].

It is generally believed that microglia utilize glycolysis and oxidative phosphorylation (OXPHOS) to meet the required energy demands for their functions under homeostatic conditions (Fig. 2A) [27, 28]. Glycolysis begins with the uptake of extracellular glucose by glucose transporters (GLUTs). In a homeostatic state, microglia predominantly express GLUT1 and GLUT5 (a fructose transporter) as the primary transmembrane sugar transporters [29, 30]. However, decreased transportation of glucose into the brain coupled with reduced oxidation rates contributes to a state referred to as ‘hypometabolism’, which is associated with AD pathology [31]. Under inflammatory conditions, GLUT1 expression is upregulated to boost glucose uptake and facilitate glycolysis in microglia [32, 33]. Biswas et al. reported a reduction.

In GLUT1 and GLUT3 expression, as well as a decrease in brain glucose concentration, in an experimental rat model of AD, which was generated by the administration of streptozotocin, an alkylating glucosamine nitrosourea compound commonly used in rodent models to induce AD [34]. These changes were accompanied by a decrease in CD11b immunoreactivity, a biomarker expressed on the surface of myeloid cells, including microglia in the brain. Early studies in the 1990s revealed a diminished concentration of GLUT1 and GLUT3 in the brains of AD patients, which was correlated with decreased brain glucose uptake and subsequent cognitive impairment [35, 36]. However, in vivo imaging of neuroinflammation via small-animal positron emission tomography (PET) revealed a positive correlation between glucose metabolism and the concomitant neuroinflammation as measured by the translocator protein (TSPO), an 18 kDa biomarker expressed in the mitochondrial membrane of activated microglia, in a transgenic AD mouse model [37]. A large proteomic study on AD, involving over 2,000 AD brain samples and 400 cerebrospinal fluid (CSF) samples from individuals with AD, revealed elevated levels of glucose metabolism-related proteins [38]. These proteins, which include astrocyte/microglial carbohydrate metabolism enzymes such as pyruvate kinase M2 (PKM2), lactate dehydrogenase (LDH)-B, and glyceraldehyde 3-phosphate dehydrogenase, were found to be highly interconnected within specific protein networks known as M4 modules [38]. In addition, the M4 module contained microglial markers that increased in response to Aβ deposition and decreased in response to lipopolysaccharide (LPS) stimulation, implying that the microglial activity reflected by the M4 module is likely inclined towards an anti-inflammatory, disease-related phenotype. While hypometabolism is recognized as a biomarker of AD, the increased expression of metabolic proteins and elevated glucose consumption in microglia has the propensity to mask the hypometabolic phenotypes of AD brains [39].

Of further interest is the emergence of studies indicating that alterations in glucose metabolism may regulate phenotype-related changes in microglia. Under normoxia (normal oxygen levels), pyruvate dehydrogenase transforms pyruvate into acetyl-CoA, which subsequently enters the tricarboxylic acid (TCA) cycle, generating reduced flavin adenine dinucleotide (FADH2) and nicotinamide adenine dinucleotide (NADH) as electron donors for electron transport chain (Fig. 2A). Alternatively, pyruvate can be preferentially converted to lactate through a process catalyzed by LDH-A. While traditionally associated with anaerobic conditions, recent evidence from immune activation demonstrates that this pathway occurs even under normoxic conditions. This process, known as aerobic glycolysis, is similar to what is observed in tumor cells and is often referred to as the Warburg effect [18, 40]. Aerobic glycolysis supports the quick production of adenosine triphosphate (ATP) and also facilitates the generation of cytokines and reactive oxygen species (ROS) in activated microglia through several ancillary pathways [41]. Maintenance of an equilibrium between OXPHOS and glycolysis is vital for orchestrating the shift between inflammatory and homeostatic functions in immune cells [42,43,44]. Baik et al. reported that in vitro exposure to Aβ plaques directly induced microglial activation by shifting OXPHOS to aerobic glycolysis, which led to a surge in proinflammatory cytokine production and an increase in the phagocytic phenotype of microglia, which depended on the mTOR-HIF-1α (hypoxia-inducible factor 1-alpha) pathway (Fig. 2A and B) [45].

Fig. 2
figure 2

A comprehensive summary of metabolic pathways and intermediates regulating microglia immunological function. (A) During glycolysis, glucose is converted to pyruvate, which either enters the TCA cycle via acetyl-CoA or is converted to lactate under low-oxygen conditions (anaerobic glycolysis) or in normoxic conditions (aerobic glycolysis; Warburg phenomenon). This metabolic switch promotes the expression of anti-inflammatory markers such as IL-10, TGFβ, CD206, and VEGFα. Inflammatory conditions disrupt the TCA cycle at stages involving IDH and SDH, resulting in citrate and succinate accumulation. Citrate enhances the production of prostaglandins, NO, and itaconate, whereas succinate stabilizes HIF-1α, increasing the expression of multiple glycolytic enzymes. Microglia in proinflammatory states favor fatty acid synthesis from acetyl Co-A, which leads to the secretion of additional proinflammatory cytokines. Conversely, anti-inflammatory microglia prefer β-oxidation of fatty acids, which converge into acetyl-CoA and upregulate the production of unsaturated fatty acids. (B) The receptor TREM2 on microglia enhances mTOR signaling, increasing microglial metabolic capacity, and promoting the transition to a mature disease-associated microglial profile. Amino acid sensing via mTORC1 triggers lysosomal activation, promoting proinflammatory responses. (C) Glutamine metabolism yields glutamate and α-ketoglutarate, supporting proinflammatory processes through the TCA cycle. Arginine metabolism results in the production of NO and cytokines, while its conversion to ornithine can modulate inflammation. (D) Tryptophan breakdown to kynurenine activates anti-inflammatory gene transcription through AhR binding

During AD progression, the metabolism of microglia shifts from OXPHOS to aerobic glycolysis [45]. This alteration leads to the accumulation of intracellular lactate, which is then transported to the extracellular space by virtue of monocarboxylate transporters (MCTs). While neurons utilize lactate as a source of fuel in the wake of decreased glucose availability, excessive lactate leads to neuronal damage by altering the acidity of the AD brain microenvironment [46]. Further, lactate stimulates the microglial release of proinflammatory cytokines, interleukin (IL)-6, tumor necrosis factor (TNF)-α, and IL-1β [47]. The glycolysis-driven lactate is transported to the cell nucleus, where it leads to histone ‘lactylation’ [48]. Indeed, histone lactylation is notably increased in the brain tissues of both AD patients and mouse models of AD [48]. In particular, histone lactylation at H4 lysine 14 (H4K12la) has been identified as the most prevalent differentially altered epigenetic marker in this context [49]. In addition, H4K12la, which is specifically upregulated in Aβ-associated microglia, enhances the activity of glycolytic genes (Pkm and Ldha). This establishes a positive feedback loop involving glycolysis, H4K12la, and PKM2, exacerbating glucose metabolism, which induces microglial dysfunction in AD pathogenesis. Intriguingly, interrupting this cycle resulted in a reduced Aβ burden, inhibited microglial activation, and improved memory and spatial learning in mice [46]. These findings suggest that targeting the glycolysis/H4K12 lactylation/PKM2 axis in microglia has potential as a therapeutic strategy for the treatment of AD.

Studies involving the triggering receptor expressed on myeloid cells-2 (TREM2) have provided additional evidence supporting the role of metabolic pathways in enhancing microglial immune responses. TREM2, an immunoglobulin-like orphan receptor of the TREM family, is essential for microglial phagocytic and apoptotic responses [50]. TREM2 augments mTOR signaling, enhances microglial metabolic capacity, and facilitates the transition of microglia to a disease-associated microglia profile, which ultimately supports the microglial response to Aβ-plaques [51, 52]. The transmembrane region of TREM2 is associated with the adaptor proteins DNAX-activation protein 12 (DAP12) and DAP10 (Fig. 2B). Consequently, TREM2 activates mTOR through DAP12 and/or DAP10, given that both signaling adaptors have the capacity to recruit upstream mTOR activators [53]. Kleinberger et al. created a mouse model of neurodegenerative disorders by introducing disease-associated Trem2p.T66M mutation (T66M) in mice, which mirrored frontotemporal dementia associated with AD [54]. PET imaging revealed that the brains of T66M mutants exhibited greater glucose utilization than those of the wild-type brains. Additionally, there was an age-dependent decrease in proinflammatory activation of microglia in these mice, as shown by Iba1 immunohistochemistry and longitudinal PET imaging of the TSPO ligand. A study by Ulland et al. demonstrated that the microglia in the TREM2-deficient mouse model of AD and AD patients who harbor TREM2 risk variants experienced aberrant autophagy, which is associated with impaired mTOR signaling [55]. Autophagy, an intracellular degradation pathway that is partly controlled by mTOR-dependent signaling, is vital for maintaining energy equilibrium and clearance of misfolded proteins and damaged organelles [56, 57]. Furthermore, compared to wild-type bone marrow-derived macrophages (BMDMs), Trem2−/− BMDMs presented a reduction in the levels of glycolytic metabolites such as glucose 6-phosphate and fructose 1,6-bisphosphate, as well as TCA cycle intermediates, including citrate and succinate. Trem2−/− BMDMs presented a decreased extracellular acidification rate, suggesting diminished glycolytic flux [55]. However, it is still unclear whether TREM2 promotes neuroprotection through its functional roles in microglia.

Enhanced lipid metabolism also provides the energy needed for microglia activation, and disruptions in this process are strongly associated with lipid metabolism disorders, such as hypercholesterolemia, hypertriglyceridemia, diabetes, and even AD [58,59,60]. Apolipoprotein E (ApoE), the primary cholesterol transporter in the brain, is widely recognized as a significant genetic risk factor for the onset of late-stage AD [61]. ApoE, together with TREM2, is implicated in AD pathogenesis, particularly in the context of microglia activation (Fig. 2B) [13]. Therefore, understanding the interactions between these two pathways may provide insights into the underlying mechanisms of AD pathogenesis and design potential therapeutic strategies. In the 5XFAD mouse model of AD, a novel disease-associated microglia was identified [51]. These microglia subsets had a unique transcriptional profile involving lipid and metabolism-related genes, including Trem2, a lipoprotein lipase, and ApoE. This subset of microglia preferentially utilized lipids as an energy source to meet their heightened metabolic demands when activated. Similarly, in both AD and APP-PS1 mouse models, the TREM2-driven ApoE pathway triggered a shift in microglial phenotypes from a homeostatic state to a neurodegenerative one, suggesting that activation of the TREM2-ApoE pathway impairs the ability of microglia to maintain homeostasis in the brain [13].

In AD, in addition to disruptions in lipid metabolism, alterations in amino acid metabolism play crucial roles in disease progression. Amino acids play vital roles in the biosynthesis of proteins, thereby influencing a range of metabolic pathways essential for cellular functions. Amino acids also support immune cell energy acquisition, biomass production, and metabolic reprogramming during activation [62]. Glutamine, which serves as an important energy source for microglia, is absorbed through the amine transporters, SLC1A5 and SLC38A1 [63]. Inside mitochondria, glutamine is converted to glutamate and ammonia by the activity of glutaminase (Fig. 2C). Glutamate is further metabolized to α-ketoglutarate by glutamate dehydrogenase 1, which then enters the TCA cycle to produce ATP. Glutamate and α-ketoglutarate eventually generate succinate via the gamma-aminobutyric acid (GABA)-shunt pathway, triggering the production of proinflammatory IL-1β by modulating HIF-1α and promoting a shift to the proinflammatory phenotype [64,65,66]. Glutaminase is, thus, upregulated in activated microglia under pathological conditions [67,68,69]. In vitro studies highlight that microglia express glutamine synthetase, which facilitates the conversion of glutamate to glutamine upon stimulation with inflammatory triggers such as LPS [70]. Furthermore, elevated levels of glutaminase in AD transgenic mouse brain tissues compared with those in littermate controls are correlated with increased levels of proinflammatory markers [67]. Tryptophan metabolism within microglia also plays a significant role in AD pathology (Fig. 2D). Tryptophan metabolites such as 5-hydroxyindole-acetic acid (5-HIAA) and kynurenic acid enhanced neprilysin expression, a key enzyme involved in the clearance of Aβ peptides in the brain [71]. Tryptophan is metabolized mainly via the kynurenine pathway, with indoleamine 2,3-dioxygenase serving as the rate-limiting enzyme [72]. This enzyme is notably upregulated in AD brains, which facilitates the conversion of tryptophan into quinolinic acid [73]. The kynurenine pathway begins with the oxidative cleavage of the indole ring of l-tryptophan by either indoleamine 2,3-dioxygenase or tryptophan 2,3-dioxygenase, leading to the formation of N-formylkynurenine [72]. This is followed by a series of metabolic reactions that ultimately generate NAD + and 3-hydroxykynurenine, which further generate highly reactive free radicals. 3-hydroxykynurenine is metabolized into neurotoxic compounds, including hydroxyanthranilic and quinolinic acids. Quinolinic acid stimulates glutamate release in microglia, triggering substantial production of ROS, lipid peroxidation, and tau protein hyperphosphorylation [71, 74, 75]. Methionine and Arginine metabolism are also of interest to the field [76,77,78,79,80]. Recent findings have shown that repeated high-dose administration of methionine enhanced microglial proinflammatory activation [76,77,78]. Further, Arginine contributed to pro- and anti-inflammatory responses through enzymes such as inducible nitric oxide synthase, arginase-1, arginine decarboxylase, and arginine-glycine amidino transferase in microglia [79, 80]. In a mouse model of AD, hippocampal neuronal death was associated with an increase in immunosuppressive CD11c + microglia and extracellular arginase, leading to arginine breakdown and decreased brain arginine levels [81]. Pharmacological blocking of arginase and ornithine decarboxylase shielded the mice from AD-like pathology and notably lowered CD11c expression [81]. Additionally, Aβ specifically triggered disease-associated microglial signatures to enhance the phagocytic response, whereas a deficiency in myeloid arginase 1 selectively induced a homeostatic, nonphagocytic microglia signature [82].

Microglia also express diverse iron-related proteins, including the transferrin receptor, divalent metal transporter-1, ferritin, and ferroportin, which play integral roles in the absorption and metabolic processes associated with iron [83,84,85]. Compared with neurons and astrocytes, microglia exhibit superior efficiency in the accumulation and storage of iron in vitro [84]. Iron-laden microglia have been identified in the hippocampus [86] and in the frontal cortex of individuals with AD and are typically concentrated in the vicinity of Aβ plaques [87]. The retention of iron within microglia is associated with elevated glycolytic activity and enhanced TNFα expression [88]. Indeed, exposure of microglia to IFNγ and Aβ peptides in vitro increased glycolysis and associated glycolytic enzymes and enhanced iron retention while concurrently diminishing phagocytic and chemotactic functions [89]. These findings indicate that inflammation driven by iron-associated microglia, particularly in conjunction with Aβ, may play a pivotal role in the process of neurodegeneration during AD pathology.

Multiple sclerosis (MS)

MS is an immune-mediated neurodegenerative disease characterized by the demyelination of axons within the CNS (Fig. 1). According to the Multiple Sclerosis International Federation report, more than 2.9 million patients suffer from MS worldwide [90]. While the etiology of MS remains incompletely understood, the immunopathophysiology of MS involves intricate interactions among various peripheral immune cells and CNS-resident cells. In the early stages of pathology, the integrity of the blood-brain barrier (BBB) is compromised, as evidenced by gadolinium-enhancing MRI reflecting active lesions within the brains of MS patients [91, 92]. The breached BBB allows the infiltration of peripheral monocytes, B cells, and autoreactive CD4 + T cells, particularly T helper (Th)1, Th17, and CD8 + T cells, into the CNS parenchyma [93,94,95,96,97]. These infiltrating cells initiate an attack on myelin within the CNS, leading to the degeneration of oligodendrocytes [98, 99], a process that involves the loss of myelin and the release of myelin debris [100]. Microglia and macrophages are essential for the phagocytic removal of inhibitory myelin debris (Fig. 2E); however, prolonged activity of microglia leads to the destruction of neuronal myelin, which eventually compromises axonal and synaptic activity [101,102,103,104].

Abnormalities in glucose metabolism, such as elevated blood glucose, increased pyruvate levels, as well as increased activity of cerebral metabolic enzymes, including enolase, pyruvate kinase, and LDH, have been implicated in the pathophysiology of MS (Fig. 2A) [105,106,107,108,109,110,111]. Currently, information on the cellular distribution of GLUTs in the brains of people with MS is limited. Nijland et al. examined brain tissue from non-neurological human controls and individuals with MS presenting chronic active lesions [112]. These lesions exhibited an inactive center surrounded by a periphery of hypertrophic microglia and macrophages, which are hallmarks of chronic active lesions [112]. The study revealed prominent expression of GLUT3 in the active rim of the lesions, notably in astrocytes. While this study found no such changes within microglia, additional sugar transporters within these cells and across the MS lesions are warranted. Immune cells that undergo excessive glycolysis witness enhanced LDHA activity. This enzyme, which converts pyruvate to lactate, helps maintain glycolytic flux as it replenishes the necessary NAD + to sustain glycolysis [113]. In this context, our previous research highlighted that CNS-infiltrating macrophages in MS brains and the brains of experimental autoimmune encephalomyelitis (EAE), a mouse model of MS, expressed high levels of LDHA and MCT-4, which enables the export of excessive lactate from macrophages [114]. The study further revealed that inhibitors of MCT-4 or knockdown of LDHA dampened macrophage activation, leading to reduced EAE severity [114]. Whether microglia exhibit similar phenotypes while contributing to MS progression remains to be identified. Nevertheless, microglia exhibit metabolic flexibility, i.e., they have the potential to utilize alternative carbon sources for energy pathways, allowing them to adapt to diverse CNS conditions. Microglia utilize glucose as a preferred metabolite to fulfill their energy demands in a healthy CNS [115]. However, they can quickly adapt to changes in the CNS microenvironment by switching to alternative pathways. In this context, a study by Bernier et al. found that, under aglycemic (no glucose) conditions in cerebellar slice cultures, microglia shifted to glutaminolysis [116]. In this process, microglia switched to using the amino acid glutamine, which was then converted into α-ketoglutarate to feed into the TCA cycle for its homeostatic functions (Fig. 2C). On the other hand, overactivation of glycolytic pathways during inflammation leads to histone acetylation and transcriptional activation of IL-1β in microglia as noted in ischemia [117]. Macrophages that undergo glycolytic activation experience interruptions during the conversion of citrate to isocitrate (crucial for the generation of nitric oxide and IL-6) and the conversion of succinate to fumarate (which stabilizes HIF-1α, leading to the generation of IL-1β) in the TCA cycle [118]. It remains to be investigated whether microglia experience similar disruptions in their metabolic program across different MS pathologies.

Given that myelin is abundant in lipids, it is not surprising that research on lipid metabolism and its role in microglial activation is gaining momentum in the MS field. 7-Keto-cholesterol (7KC) and 7β-hydroxycholesterol (7βHC) are cholesterol metabolites that serve as biomarkers of oxidative stress and are implicated in neuroinflammation [119, 120]. 7KC and 7βHC generated through ROS-mediated cholesterol oxidation are linked to neuroaxonal injury, as measured by the serum neurofilament light chain within the serum of MS patients [121]. In addition, the high concentrations of 7KC detected in the CSF of newly diagnosed MS patients have the potential to induce neuronal damage through microglia activation [122]. 7KC enters the cellular nuclei and activates poly (ADP‒ribose)-polymerase, which subsequently induces a proinflammatory phenotype characterized by the expression of integrins, such as CD11a, and intercellular adhesion molecules [122, 123]. In contrast, polyunsaturated fatty acids (PUFAs) induce an anti-inflammatory phenotype in microglia, as increased consumption of total dietary PUFAs has been linked to a decreased risk of developing MS [124, 125]. A recent study noted that a ketogenic diet in a mouse model of MS suppressed microglial activation and attenuated neuroinflammation in the spinal cord through the regulation of the NF-κB/NLRP3 pathway and the inhibition of histone deacetylase 3 and P2 × 7R functions [126]. Another study noted elevated soluble TREM2 levels in the CSF of MS patients, where TREM2 was also found to be enriched within the lipid-laden microglia and infiltrating monocytes in actively demyelinating lesions [127]. While the conditional knockout of TREM2 in CD4 + T cells resulted in a significant reduction in CNS infiltration of Th17 cells and the production of inflammatory cytokines in EAE, the role of TREM2 in microglia has also been the focus of several recent studies [128]. For instance, the toxic oxidized phosphatidylcholines, which accumulated in MS lesions and an animal model of demyelination, TREM2 was found to be important for its clearance by microglia [129]. Also, TREM2 expression increased in microglia during EAE [130], and its inhibition during the effector phase of EAE was associated with widespread inflammation and demyelination in the brain [131]. This is likely due to a prominent role for TREM2 as a metabolic sensor that helps microglia to phagocytose myelin debris to create an environment that supports remyelination (Fig. 2B).

Similar to CNS conditions, amino acid metabolism is intricately linked to the pathophysiology of MS, affecting both immune responses and regenerative processes within the CNS (Fig. 2C, D) [132, 133]. Mass spectrometry analysis of demyelinated lesions showed a notable increase in amino acid levels during the early post-demyelination event, followed by a gradual decline during the later stages of remyelination [134]. Depending on the availability of amino acids and the activity of enzymes involved in their metabolism, microglia may either attain a proinflammatory phenotype, which impedes remyelination or shift to a pro-regenerative state that facilitates myelin repair. For example, a recent study showed that aromatic alpha-keto acids produced by the amino acid oxidase IL4I1 promoted efficient remyelination in lysophosphatidylcholine (LPC) demyelinated spinal cord lesions in mice [135]. Myeloid cells increased the expression of IL4I1 during remyelination, and knocking out IL4I1 in these cells led to unresolved macrophage/microglia-associated inflammation, resulting in impaired remyelination in the lesions. In another notable study, Fitzgerald et al. reported that circulating aromatic amino acid metabolites in CSF and blood samples from MS patients were strongly associated with high disability scores [136]. These metabolic abnormalities in aromatic amino acid metabolism resulted in an imbalance in the production of immunomodulatory pathways (e.g., hydroxycarboxylic acid-3 receptor agonists and aryl hydrocarbon receptor), particularly in a subset of CSF-enriched monocytes with a gene signature which closely resembled homeostatic microglia.

Metabolic changes in microglia are particularly evident in patients with progressive MS. Gene network analysis of brain samples from progressive MS patients revealed alterations in genes related to glycolysis and metal ion homeostasis within normal-appearing gray matter microglia in MS [137]. Another distinctive feature of chronic active MS lesions is the significant enrichment of iron within activated microglia at the rim of chronic active white matter demyelinating lesions [138]. Iron accumulation has been observed in MS histologically and via MRI, and studies have demonstrated that elevated levels of iron in the brain induced oxidative stress, potentially leading to neurodegeneration [139]. Another single-nucleus RNA sequencing study conducted on the postmortem MS brain cortices revealed a coupling of HIF-1α and the iron-induced responses in at least one subset of microglia [140]. These findings imply that alterations in iron metabolism result in oxidative stress, which triggers inflammatory pathways in microglial cells. Such observations align with other neurodegenerative disorders, such as AD [141]. On the other hand, microglia in the normal-appearing white matter (NAWM) of MS brains exhibit enrichment in genes related to lysosomal pathway, lipid catabolism, and foam cell differentiation, indicating early lipid processing [137]. Moreover, PET imaging of TSPO was enhanced in the NAWM of SPMS patients [142]. Despite these observations, our understanding of microglial metabolism in the context of MS remains limited. This is partly because most analyses conducted on postmortem MS brains fail to account for the complexity and diversity of distinct cellular phenotypes.

Parkinson’s disease (PD)

As the second most prevalent neurodegenerative disorder, PD affects approximately 1% of the population aged over 60 years [143]. A genetic mutation is present in 5–10% of patients, while the etiology of the disease is unknown in the majority of cases [144]. PD is characterized by the degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the aberrant accumulation of misfolded α-synuclein (α-SYN) protein, all of which lead to movement abnormalities (Fig. 1) [145]. Microglia play a crucial role in PD pathophysiology, where they are activated by α-SYN oligomers and proinflammatory signals released from degenerating neurons [146]. Microglia actively participate in the uptake and clearance of α-SYN oligomers, which are secreted from neurons [147,148,149,150,151]. Notably, phagocytosis of neurons by microglia is evidenced by the accumulation of neuromelanin within microglia, a synaptic engulfment mechanism analogous to the Aβ-positive synapse engulfment observed in AD [152, 153]. However, phenotypic changes over time likely interfere with the ability of microglia to maintain homeostasis and neuronal support. Consequently, the presence of hypertrophic and ‘activated’ microglia is strongly associated with irreversible neurodegenerative processes in PD [154].

The intricate interplay between microglial activation and altered glucose metabolism in PD remains a subject of investigation. Activated microglia release proinflammatory cytokines and generate excessive ROS, thereby creating a neuroinflammatory milieu that has the potential to directly impact neighboring neurons by targeting neuronal metabolism [155]. Concomitantly, PET studies revealed decreased brain glucose metabolism in the initial stages of PD [156], which correlated with increased microglial activation in the cortex and basal ganglia of PD patients [157,158,159]. Further investigations revealed substantial upregulation of Slc2A1 transcript levels (GLUT1 gene) and enhanced glycolysis as manifested by increased lactate production in primary microglia cultures exposed to α-SYN [160, 161]. Interestingly, recombinant α-SYN also enhanced glucose uptake both in the adipose tissues and skeletal muscles of mice as well as in cultured mouse preadipocytes through the PI3K/Akt pathway [162]. Acute stimulation of microglia by prefabricated α-SYN fibers in a PD mouse model resulted in a metabolic shift from OXPHOS to aerobic glycolysis through the AKT–mTOR–HIF-1α pathway in these cells (Fig. 2A) [163]. While α-SYN enhances aerobic glycolysis through pyruvate kinase M2 and suppresses mitochondrial biogenesis and OXPHOS [160], silencing of α-SYN significantly downregulates HIF-1α in microglia [164]. Notably, the mTOR/HIF-1α and ROS/HIF-1α pathways play important roles in the induction of aerobic glycolysis, which likely occurs during the Cdc-like kinase 1 (CLK1) deficiency. Clk1, also known as Coq7, encodes a mitochondrial hydroxylase essential for the biosynthesis of ubiquinone (UQ or coenzyme Q), which is crucial for the electron transport system within the mitochondrial respiratory chain and contributes to antioxidant activity [165]. The activation of these signaling pathways due to CLK1 deficiency reduced dopaminergic neurons in a mouse model of PD [166]. This investigation provided additional validation that HIF-1α assists with metabolic reprogramming to promote subsequent inflammatory responses by microglia. Increased glycolysis coincides with enhanced production of ROS in proinflammatory microglia [167]. ROS production, in turn, activates the NF-κB pathway, which serves as an inflammatory cascade capable of escalating neuroinflammation, ultimately contributing to PD pathogenesis [168,169,170].

Studies have also investigated the role of amino acid metabolism in microglia during PD pathology. Activated microglia have the potential to release substantial amounts of glutamate, leading to excitoneurotoxicity and subsequent neuronal damage in neurodegenerative disorders, including PD (Fig. 2C) [171]. Furthermore, stimulation of microglia by α-SYN led to a reduction in the level of intracellular glutamate while increasing extracellular concentrations. This modulation is believed to impact the glutamate-glutamine cycle, a crucial cycle for bioenergetic homeostasis and, therefore, neurotransmission within the neurons [172, 173]. Moreover, in Park7 gene knockout mice, which lack the protein deglycase DJ-1 and exhibit PD pathology, microglia undergo significant alterations in their central amino acid metabolism [174]. This leads to a reduction in glutamine influx and serine biosynthesis, which ultimately enhances the susceptibility of dopaminergic neurons to early cell death.

Histological examination of brain specimens revealed that microglia significantly accumulated intracellular lipids in the substantia nigra of PD patients [175], which likely fosters inflammatory processes [176, 177]. Despite these intriguing observations, the role of microglial lipid metabolism in mediating PD pathogenesis remains poorly defined.

Huntington’s disease (HD)

HD is a fatal autosomal-dominant genetic disease of the brain in which a single defective gene on chromosome 4 results in an expanded stretch of polyglutamine close to the amino terminus of the HD protein, huntingtin [178]. Consequently, pathological changes in multiple brain regions lead to a progressive disease course which is associated with a broad range of cognitive and movement disorders (Fig. 1). Recent literature suggests that microglia upregulate the production of proinflammatory cytokines, which strongly contributes to the chronic inflammation associated with HD [179, 180].

In HD brains, microglia engage in diverse signaling pathways- the kynurenine pathway, cannabinoid receptor pathway, and NF-κB signaling [181,182,183,184]. Mutant huntingtin stimulates NF-κB signaling, which contributes to the sustained activation of microglia as they secrete proinflammatory cytokines such as TNF-α [181]. This leads to chronic inflammation manifested by oxidative stress, mitochondrial dysfunction, and neuronal death, all of which drive neurodegeneration in HD [185,186,187,188]. Therefore, modulation of inflammatory response should mitigate neurotoxicity associated with mutant huntingtin [189]. In this regard, treatment of primary mouse microglia with deoxy-D-glucose, an inhibitor of glycolysis, resulted in a reduction in TNFα and IL-6 production, which was attributed to NF-κB inhibition [190]. Further, the kynurenine pathway is responsible for more than 95% of tryptophan metabolism in mammals (Fig. 2D) [191]. The activation of microglia and the subsequent neuroinflammatory response in HD patients resulted in increased levels of downstream kynurenine pathway metabolites. Mutant huntingtin fragments also activate the kynurenine pathway in microglia, which is ameliorated by treatment with histone deacetylase inhibitors in a mouse model of HD [192]. Moreover, L-kynurenine, a substrate of the kynurenine pathway, exerted a dose-dependent inhibitory effect on the LPS response within microglia [72].

Treatment with pyruvate has neuroprotective effects on striatal neurons through blocking the expression of inducible nitric oxide synthase and nitrotyrosine in activated microglia [193, 194]. Further, there was a significant increase in iron levels in the basal ganglia in symptomatic and asymptomatic HD patients [195, 196]. Using the human brain samples and the R6/2 transgenic mouse model of HD, Simmons et al. reported that disturbances in iron metabolism in HD are linked primarily to microglia and occur early enough to contribute to the progression of HD [197]. Further, dysregulation of lipid metabolism, particularly brain cholesterol metabolism, is another important feature of HD brains [198]. Despite this information, research on the specific role of lipid and iron metabolism in microglia related to HD is limited. Gaining insight into these cellular mechanisms within microglia is essential for developing therapies that could slow HD progression and improve patient outcomes.

Amyotrophic lateral sclerosis (ALS)

ALS, commonly referred to as Lou Gehrig’s disease or motor-neuron disease, is a progressive neurodegenerative disorder that affects motor neurons in both the brain and the spinal cord (Fig. 1). While the majority of ALS cases are sporadic, approximately 10% of ALS cases exhibit a familial pattern, implicating a genetic component in the disease’s etiology [199]. Mutations in genes such as superoxide dismutase 1 (SOD1) lead to the misfolding of proteins, resulting in the formation of toxic aggregates within neurons [200]. These protein aggregates interfere with cellular processes and cause susceptibility of motor neurons in ALS. Microglia play dual roles in ALS pathology, as they exhibit neuroprotective functions by assisting in the clearance of cellular debris and promoting tissue repair while also contributing to a proinflammatory state that causes neuronal damage as a result of persistent activation [201, 202].

Metabolic dysfunction is increasingly recognized as a contributing factor to ALS progression. High metabolic levels at rest, abnormalities in energy metabolism, and mitochondrial dysfunction have been observed in both neurons and glial cells in ALS patients [203,204,205]. Aerobic glycolysis, which is believed to occur through the activation of GLUTs, pyruvate dehydrogenase kinase 1, PKM2, LDH-A, and MCT-1 and inactivation of the pyruvate dehydrogenase complex, is associated with ALS pathology (Fig. 2A) [206]. Notably, disruption of metabolic support for neurons, including impaired glucose uptake and utilization, likely results from altered microglial phenotypes in individuals with ALS [204]. This contributes to the energy imbalance as observed in the affected neurons [204, 205]. In the context of ALS, TREM2-induced ApoE signaling plays a critical role (Fig. 2B), as targeting the TREM2-ApoE pathway restored the homeostatic signatures of disease-associated microglia and prevented neuronal loss in ALS mouse models [13]. Furthermore, mitochondria harvested from crude spinal cord homogenates in individuals with ALS reflected a dysfunction in electron transport chain complexes I + III, II + III, and IV [207]. Moreover, a reduction in the activity of mitochondrial enzymes, citrate synthase, and cytochrome c oxidase was observed in the motor neurons of individuals affected by ALS [208]. Proinflammatory signals from microglia likely affect mitochondrial dynamics, leading to impaired energy production and increased oxidative stress [209]. This, in turn, contributes to the overall metabolic imbalance observed in ALS. On the other hand, Olesoxime, a novel neuroprotective compound believed to stabilize mitochondrial functions, was shown to decrease the activation of microglia and delay muscle denervation and motor neuron death in an ALS mouse model [210]. Studies have also focused on the role of amino acid metabolism in ALS. Niida-Kawaguchi et al. discovered that microglia increase glutamate release in the ALS spinal cord [211], which results from the buildup of intracellular iron. This accumulation activates aconitase 1, a TCA cycle enzyme, along with the TNF-alpha converting enzyme (Fig. 2C). This process also promotes upregulation of glutaminase C, which is stimulated by extracellular TNFα. In addition, group I metabotropic glutamate receptors (mGluRs) on microglia have immunoprotective functions. Suppressing mGluR1 or mGluR5 in ALS animal models has been shown to delay the onset of the disease, extend survival, protect neurons, and decrease microglial activation [212,213,214]. These results emphasize the intricate relationship between metabolic disturbances and neuroinflammation in ALS, pointing to the importance of exploring metabolic treatments for managing the disease. Ongoing research into the metabolic pathways in ALS could lead to new therapeutic approaches that help protect neurons and improve patient survival.

Table 1 provides a broad overview of how different metabolic pathways, including glycolysis, lipid metabolism, and iron metabolism, affect neurodegenerative diseases in distinct ways.

Table 1 Summary of key metabolic processes that guide microglia function in neurodegenerative diseases

Conclusion

Metabolic reprogramming significantly influences microglial functions and emerges as a common theme across different neurodegenerative disorders. One of the key points of discussion in this review is the central position that glycolytic pathways assume in promoting the proinflammatory phenotype of activated microglia. The involvement of glucose metabolism in microglia is intricately linked to the complex mechanisms underlying neuroinflammation. Studies have shown that inhibiting glycolysis in microglia suppresses their proinflammatory response and reduces neuroinflammation [27, 226]. Therefore, targeting glucose metabolism in microglia represents a promising therapeutic approach for the treatment of debilitating CNS disorders. One such potential therapeutic target is the enzyme hexokinase 1, which catalyzes the first rate-limiting step of glycolysis by phosphorylating glucose to produce glucose-6-phosphate. The inhibition of hexokinase 1 has been shown to promote debris clearance by microglia while it reduced neuroinflammation in animal models [215, 218]. Another promising target is the enzyme pyruvate dehydrogenase kinase, which regulates the activity of the pyruvate dehydrogenase complex and thus controls the entry of pyruvate into the TCA cycle. Inhibition of pyruvate dehydrogenase kinase is a focal point in cancer research, but its impact on promoting anti-inflammatory phenotypes within microglia in degenerative disorders remains understudied [227]. In addition to discussing the relevance of glycolysis, the role of fatty acid metabolism in microglia has also been highlighted in this review. Fatty acid oxidation is essential for the anti-inflammatory phenotype of microglia, and enhancing this metabolic pathway may offer therapeutic benefits for neuroinflammatory diseases. For example, the peroxisome proliferator-activated receptor is a crucial transcription factor that facilitates the transfer of fatty acids into the mitochondria for oxidation and might play a neuroprotective role by inhibiting microglia activation [228,229,230].

While numerous pathways have been discussed in this review, ascribing metabolic pathways to distinct microglia populations presents significant challenges. Microglia represent a heterogeneous population of cells with distinct metabolic profiles depending on their location, microenvironment, and activation states [137, 231]. Additionally, metabolic pathways are tightly linked to diverse functions of microglia, including immune responses, phagocytosis, and synaptic pruning. For example, interruption in glycolysis reduces TNFα and IL-6 production through the inhibition of NF-kB pathway within primary microglia [190, 232]. Further, glia-derived IL-33 enhanced mitochondrial activity and phagocytosis in microglia via the AKT-dependent pathway, facilitating synapse engulfment during neurodevelopment, while disruption of the IL-33-AKT signaling axis impaired metabolic adaptation and phagocytic functions, hindering neurodevelopment [233]. Therefore, targeting metabolic pathways may impact different subsets of microglia in various ways, complicating therapeutic strategies. More importantly, modulating metabolic pathways in microglia may have off-target effects, potentially leading to systemic dysfunction or disrupting physiological processes outside the CNS. On the other hand, microglia adapt their metabolic profile in response to changes in the microenvironment or disease conditions [234]. Therefore, targeting a single metabolic pathway might induce compensatory changes in other subsets, potentially negating the effect of therapies. Moreover, approaches to address metabolic alterations in microglia should involve compounds that are both low in molecular weight and lipophilic to ensure they can effectively cross the BBB [235]. Neurodegenerative diseases like MS, AD, PD, HD, and ALS involve intricate pathophysiological mechanisms with multiple contributing factors. As a result, strategies aimed at targeting microglial metabolism may need to be customized to specific disease stages and contexts, adding to the complexity of therapeutic development.

In summary, therapeutic modulation of metabolic processes in microglia represents a promising approach for developing novel treatments for neurodegenerative diseases. By understanding and manipulating microglial metabolic regulation, we can potentially harness their neuroprotective abilities, offering new hope for patients with severe neurological conditions. Nevertheless, additional research is required to thoroughly understand the intricate metabolic regulation of microglial functions and to determine the most effective strategies for targeting these processes.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

5-HIAA:

5-hydroxyindole-acetic acid

7βHC:

7β-hydroxycholesterol

7KC:

Keto-cholesterol

Aβ:

Amyloid-β

AD:

Alzheimer’s disease

ALS:

Amyotrophic lateral sclerosis

ApoE:

Apolipoprotein E

ATP:

Adenosine triphosphate

BMDMs:

Bone marrow-derived macrophages

CD:

Cluster of Differentiation

CLK1:

Cdc-like kinase 1

CNS:

Central nervous system

CSF:

Cerebrospinal fluid

DAP12:

DNAX-activation protein 12

EAE:

Experimental autoimmune encephalomyelitis

FADH2:

Flavin adenine dinucleotide

G6P:

Glucose-6-phosphate

GABA:

Gamma-aminobutyric acid

GDP:

Guanosine diphosphate

GLUT:

Glucose transporter

H4K12la:

Histone lactylation at H4 lysine 14

HD:

Huntington’s disease

HDAC3:

Histone deacetylase 3

HIF-1α:

Hypoxia-inducible factor 1-alpha

IL:

Interleukin

LDH:

Lactate dehydrogenase

LPC:

Lysophosphatidylcholine

LPS:

Lipopolysaccharide

mGluRs:

Metabotropic glutamate receptors

MCT:

Monocarboxylate transporter

mRNA:

Messenger ribonucleic acid

MS:

Multiple sclerosis

mTOR:

Mechanistic target of rapamycin

NAD+:

Nicotinamide adenine dinucleotide

NADH:

Nicotinamide adenine dinucleotide (reduced form)

NAWM:

Normal-appearing white matter

NF-κB:

Nuclear factor kappa-light-chain-enhancer of activated B cells

NLRP3:

NLR family pyrin domain containing 3

NO:

Nitric oxide

OXPHOS:

Oxidative phosphorylation

PD:

Parkinson’s disease

PET:

Positron emission tomography

PI3Ks:

Phosphoinositide 3-kinases

PKM2:

Pyruvate kinase M2

PUFAs:

Polyunsaturated fatty acids

ROS:

Reactive oxygen species

SDH:

Succinate dehydrogenase

SNpc:

Substantia nigra pars compacta

SOD1:

Superoxide dismutase 1

SYN:

Synuclein

TCA:

Tricarboxylic acid

Th:

T helper

TNF:

Tumor necrosis factor

TNFα:

Tumor necrosis factor-alpha

TREM:

Triggering receptor expressed on myeloid cells

α-SYN:

α-synuclein

References

  1. Kierdorf K, Erny D, Goldmann T, Sander V, Schulz C, Perdiguero EG, et al. Microglia emerge from erythromyeloid precursors via Pu. 1-and Irf8-dependent pathways. Nat Neurosci. 2013;16(3):273–80.

    Article  CAS  PubMed  Google Scholar 

  2. Perry V, Hume DA, Gordon S. Immunohistochemical localization of macrophages and microglia in the adult and developing mouse brain. Neuroscience. 1985;15(2):313–26.

    Article  CAS  PubMed  Google Scholar 

  3. Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science. 2005;308(5726):1314–8.

    Article  CAS  PubMed  Google Scholar 

  4. Ransohoff RM, Perry VH. Microglial physiology: unique stimuli, specialized responses. Annu Rev Immunol. 2009;27:119–45.

    Article  CAS  PubMed  Google Scholar 

  5. Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, et al. ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci. 2005;8(6):752–8.

    Article  CAS  PubMed  Google Scholar 

  6. Hickman SE, Kingery ND, Ohsumi TK, Borowsky ML, Wang L-c, Means TK, et al. The microglial sensome revealed by direct RNA sequencing. Nat Neurosci. 2013;16(12):1896–905.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Perry VH, Nicoll JA, Holmes C. Microglia in neurodegenerative disease. Nat Reviews Neurol. 2010;6(4):193–201.

    Article  Google Scholar 

  8. Agrawal M. Molecular basis of chronic neurodegeneration. Clinical molecular medicine: Elsevier; 2020. pp. 447–60.

    Google Scholar 

  9. Erkkinen MG, Kim M-O, Geschwind MD. Clinical neurology and epidemiology of the major neurodegenerative diseases. Cold Spring Harb Perspect Biol. 2018;10(4):a033118.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Nichols E, Szoeke CE, Vollset SE, Abbasi N, Abd-Allah F, Abdela J, et al. Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990–2016: a systematic analysis for the global burden of Disease Study 2016. Lancet Neurol. 2019;18(1):88–106.

    Article  Google Scholar 

  11. Hou Y, Dan X, Babbar M, Wei Y, Hasselbalch SG, Croteau DL, et al. Ageing as a risk factor for neurodegenerative disease. Nat Reviews Neurol. 2019;15(10):565–81.

    Article  Google Scholar 

  12. Navarro V, Sanchez-Mejias E, Jimenez S, Muñoz-Castro C, Sanchez-Varo R, Davila JC, et al. Microglia in Alzheimer’s disease: activated, dysfunctional or degenerative. Front Aging Neurosci. 2018;10:357752.

    Article  Google Scholar 

  13. Krasemann S, Madore C, Cialic R, Baufeld C, Calcagno N, El Fatimy R, et al. The TREM2-APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. Immunity. 2017;47(3):566–81. e9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Yong VW. Microglia in multiple sclerosis: protectors turn destroyers. Neuron. 2022;110(21):3534–48.

    Article  CAS  PubMed  Google Scholar 

  15. Afridi R, Lee W-H, Suk K. Microglia gone awry: linking immunometabolism to neurodegeneration. Front Cell Neurosci. 2020;14:246.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lynch MA. Can the emerging field of immunometabolism provide insights into neuroinflammation? Prog Neurobiol. 2020;184:101719.

    Article  CAS  PubMed  Google Scholar 

  17. Benarroch E. What is the role of Microglial metabolism in inflammation and neurodegeneration? Neurology. 2022;99(3):99–105.

    Article  PubMed  Google Scholar 

  18. Kaushik DK, Yong VW. Metabolic needs of brain-infiltrating leukocytes and microglia in multiple sclerosis. J Neurochem. 2021;158(1):14–24.

    Article  CAS  PubMed  Google Scholar 

  19. Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991;82(4):239–59.

    Article  CAS  PubMed  Google Scholar 

  20. Hardy JA, Higgins GA. Alzheimer’s disease: the amyloid cascade hypothesis. Science. 1992;256(5054):184–5.

    Article  CAS  PubMed  Google Scholar 

  21. Lai AY, McLaurin J. Clearance of amyloid-β peptides by microglia and macrophages: the issue of what, when and where. Future Neurol. 2012;7(2):165–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Pereira JB, Janelidze S, Strandberg O, Whelan CD, Zetterberg H, Blennow K, et al. Microglial activation protects against accumulation of tau aggregates in nondemented individuals with underlying Alzheimer’s disease pathology. Nat Aging. 2022;2(12):1138–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Condello C, Yuan P, Schain A, Grutzendler J. Microglia constitute a barrier that prevents neurotoxic protofibrillar Aβ42 hotspots around plaques. Nat Commun. 2015;6(1):6176.

    Article  CAS  PubMed  Google Scholar 

  24. Ising C, Venegas C, Zhang S, Scheiblich H, Schmidt SV, Vieira-Saecker A, et al. NLRP3 inflammasome activation drives tau pathology. Nature. 2019;575(7784):669–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, et al. NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature. 2013;493(7434):674–8.

    Article  CAS  PubMed  Google Scholar 

  26. Gordon R, Albornoz EA, Christie DC, Langley MR, Kumar V, Mantovani S, et al. Inflammasome inhibition prevents α-synuclein pathology and dopaminergic neurodegeneration in mice. Sci Transl Med. 2018;10(465):eaah4066.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Cheng J, Zhang R, Xu Z, Ke Y, Sun R, Yang H, et al. Early glycolytic reprogramming controls microglial inflammatory activation. J Neuroinflamm. 2021;18(1):129.

    Article  CAS  Google Scholar 

  28. Zhang Y, Chen K, Sloan SA, Bennett ML, Scholze AR, O’Keeffe S, et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J Neurosci. 2014;34(36):11929–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Payne J, Maher F, Simpson I, Mattice L, Davies P. Glucose transporter glut 5 expression in microglial cells. Glia. 1997;21(3):327–31.

    Article  CAS  PubMed  Google Scholar 

  30. Wang L, Pavlou S, Du X, Bhuckory M, Xu H, Chen M. Glucose transporter 1 critically controls microglial activation through facilitating glycolysis. Mol Neurodegeneration. 2019;14:1–15.

    Article  Google Scholar 

  31. Camandola S, Mattson MP. Brain metabolism in health, aging, and neurodegeneration. EMBO J. 2017;36(11):1474–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Wang L, Pavlou S, Du X, Bhuckory M, Xu H, Chen M. Glucose transporter 1 critically controls microglial activation through facilitating glycolysis. Mol Neurodegeneration. 2019;14(1):1–15.

    Article  Google Scholar 

  33. Kalsbeek MJ, Mulder L, Yi C-X. Microglia energy metabolism in metabolic disorder. Mol Cell Endocrinol. 2016;438:27–35.

    Article  CAS  PubMed  Google Scholar 

  34. Biswas J, Gupta S, Verma DK, Gupta P, Singh A, Tiwari S, et al. Involvement of glucose related energy crisis and endoplasmic reticulum stress: insinuation of streptozotocin induced Alzheimer’s like pathology. Cell Signal. 2018;42:211–26.

    Article  CAS  PubMed  Google Scholar 

  35. Harr SD, Simonian NA, Hyman BT. Functional alterations in Alzheimer’s disease: decreased glucose transporter 3 immunoreactivity in the perforant pathway terminal zone. J Neuropathology Experimental Neurol. 1995;54(1):38–41.

    Article  CAS  Google Scholar 

  36. Simpson IA, Chundu KR, Davies-Hill T, Honer WG, Davies P. Decreased concentrations of GLUT1 and GLUT3 glucose transporters in the brains of patients with Alzheimer’s disease. Annals Neurology: Official J Am Neurol Association Child Neurol Soc. 1994;35(5):546–51.

    Article  CAS  Google Scholar 

  37. Brendel M, Probst F, Jaworska A, Overhoff F, Korzhova V, Albert NL, et al. Glial activation and glucose metabolism in a transgenic amyloid mouse model: a triple-tracer PET study. J Nucl Med. 2016;57(6):954–60.

    Article  CAS  PubMed  Google Scholar 

  38. Johnson EC, Dammer EB, Duong DM, Ping L, Zhou M, Yin L, et al. Large-scale proteomic analysis of Alzheimer’s disease brain and cerebrospinal fluid reveals early changes in energy metabolism associated with microglia and astrocyte activation. Nat Med. 2020;26(5):769–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Backes H, Walberer M, Ladwig A, Rueger MA, Neumaier B, Endepols H, et al. Glucose consumption of inflammatory cells masks metabolic deficits in the brain. NeuroImage. 2016;128:54–62.

    Article  CAS  PubMed  Google Scholar 

  40. Warburg O. The metabolism of carcinoma cells. J Cancer Res. 1925;9(1):148–63.

    Article  CAS  Google Scholar 

  41. Schuster S, Boley D, Möller P, Stark H, Kaleta C. Mathematical models for explaining the Warburg effect: a review focussed on ATP and biomass production. Biochem Soc Trans. 2015;43(6):1187–94.

    Article  CAS  PubMed  Google Scholar 

  42. Pålsson-McDermott EM, O’Neill LA. Targeting immunometabolism as an anti-inflammatory strategy. Cell Res. 2020;30(4):300–14.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Liang X, Wang Z, Dai Z, Zhang H, Zhang J, Luo P, et al. Glioblastoma glycolytic signature predicts unfavorable prognosis, immunological heterogeneity, and ENO1 promotes microglia M2 polarization and cancer cell malignancy. Cancer Gene Ther. 2023;30(3):481–96.

    CAS  PubMed  Google Scholar 

  44. Bernier L-P, York EM, MacVicar BA. Immunometabolism in the brain: how metabolism shapes microglial function. Trends Neurosci. 2020;43(11):854–69.

    Article  CAS  PubMed  Google Scholar 

  45. Baik SH, Kang S, Lee W, Choi H, Chung S, Kim J-I, et al. A breakdown in metabolic reprogramming causes microglia dysfunction in Alzheimer’s disease. Cell Metabol. 2019;30(3):493–507. e6.

    Article  CAS  Google Scholar 

  46. Zhao Y, Xu H. Microglial lactate metabolism as a potential therapeutic target for Alzheimer’s disease. Mol Neurodegeneration. 2022;17(1):1–3.

    Article  Google Scholar 

  47. Andersson AK, Rönnbäck L, Hansson E. Lactate induces tumour necrosis factor-α, interleukin‐6 and interleukin‐1β release in microglial‐and astroglial‐enriched primary cultures. J Neurochem. 2005;93(5):1327–33.

    Article  CAS  PubMed  Google Scholar 

  48. Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y, et al. Metabolic regulation of gene expression by histone lactylation. Nature. 2019;574(7779):575–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Pan R-Y, He L, Zhang J, Liu X, Liao Y, Gao J, et al. Positive feedback regulation of microglial glucose metabolism by histone H4 lysine 12 lactylation in Alzheimer’s disease. Cell Metabol. 2022;34(4):634–48. e6.

    Article  CAS  Google Scholar 

  50. Hsieh CL, Koike M, Spusta SC, Niemi EC, Yenari M, Nakamura MC, et al. A role for TREM2 ligands in the phagocytosis of apoptotic neuronal cells by microglia. J Neurochem. 2009;109(4):1144–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Keren-Shaul H, Spinrad A, Weiner A, Matcovitch-Natan O, Dvir-Szternfeld R, Ulland TK, et al. A unique microglia type associated with restricting development of Alzheimer’s disease. Cell. 2017;169(7):1276–90. e17.

    Article  CAS  PubMed  Google Scholar 

  52. Deczkowska A, Keren-Shaul H, Weiner A, Colonna M, Schwartz M, Amit I. Disease-associated microglia: a universal immune sensor of neurodegeneration. Cell. 2018;173(5):1073–81.

    Article  CAS  PubMed  Google Scholar 

  53. Xing J, Titus AR, Beth Humphrey M. The TREM2-DAP12 signaling pathway in Nasu–Hakola disease: a molecular genetics perspective. Res Rep Biochem. 2015:89–100.

  54. Kleinberger G, Brendel M, Mracsko E, Wefers B, Groeneweg L, Xiang X, et al. The FTD-like syndrome causing TREM 2 T66M mutation impairs microglia function, brain perfusion, and glucose metabolism. EMBO J. 2017;36(13):1837–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Ulland TK, Song WM, Huang SC-C, Ulrich JD, Sergushichev A, Beatty WL, et al. TREM2 maintains microglial metabolic fitness in Alzheimer’s disease. Cell. 2017;170(4):649–63. e13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism, and disease. Cell. 2017;168(6):960–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Galluzzi L, Pietrocola F, Levine B, Kroemer G. Metabolic control of autophagy. Cell. 2014;159(6):1263–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Loving BA, Bruce KD. Lipid and lipoprotein metabolism in microglia. Front Physiol. 2020;11:526534.

    Article  Google Scholar 

  59. Chausse B, Kakimoto PA, Kann O, editors. Microglia and lipids: how metabolism controls brain innate immunity. Seminars in cell & developmental biology. Elsevier; 2021.

  60. Fitz NF, Nam KN, Wolfe CM, Letronne F, Playso BE, Iordanova BE, et al. Phospholipids of APOE lipoproteins activate microglia in an isoform-specific manner in preclinical models of Alzheimer’s disease. Nat Commun. 2021;12(1):3416.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small G, et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science. 1993;261(5123):921–3.

    Article  CAS  PubMed  Google Scholar 

  62. Kelly B, Pearce EL. Amino assets: how amino acids support immunity. Cell Metabol. 2020;32(2):154–75.

    Article  CAS  Google Scholar 

  63. Zhang D, Hua Z, Li Z. The role of glutamate and glutamine metabolism and related transporters in nerve cells. CNS Neurosci Ther. 2024;30(2):e14617.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Vazquez-Valls E, Flores-Soto M, Chaparro-Huerta V, Torres-Mendoza B, Gudiño-Cabrera G, Rivera-Cervantes M, et al. HIF-1α expression in the hippocampus and peripheral macrophages after glutamate-induced excitotoxicity. J Neuroimmunol. 2011;238(1–2):12–8.

    Article  CAS  PubMed  Google Scholar 

  65. Tannahill G, Curtis A, Adamik J, Palsson-McDermott E, McGettrick A, Goel G, et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature. 2013;496(7444):238–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Lukyanova L, Kirova YI, Germanova E. The role of succinate in regulation of immediate HIF-1α expression in hypoxia. Bull Exp Biol Med. 2018;164:298–303.

    Article  CAS  PubMed  Google Scholar 

  67. Gao G, Zhao S, Xia X, Li C, Li C, Ji C, et al. Glutaminase C regulates microglial activation and pro-inflammatory exosome release: relevance to the pathogenesis of Alzheimer’s disease. Front Cell Neurosci. 2019;13:264.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Gao G, Li C, Zhu J, Wang Y, Huang Y, Zhao S, et al. Glutaminase 1 regulates neuroinflammation after cerebral ischemia through enhancing microglial activation and pro-inflammatory exosome release. Front Immunol. 2020;11:161.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Kono M, Yoshida N, Maeda K, Suárez-Fueyo A, Kyttaris VC, Tsokos GC. Glutaminase 1 inhibition reduces glycolysis and ameliorates lupus‐like disease in MRL/lpr mice and experimental autoimmune encephalomyelitis. Arthritis Rheumatol. 2019;71(11):1869–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Nakajima K, Kanamatsu T, Takezawa Y, Kohsaka S. Up-regulation of glutamine synthesis in microglia activated with endotoxin. Neurosci Lett. 2015;591:99–104.

    Article  CAS  PubMed  Google Scholar 

  71. Savonije K, Weaver DF. The role of tryptophan metabolism in alzheimer’s disease. Brain Sci. 2023;13(2):292.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Maddison DC, Giorgini F, editors. The kynurenine pathway and neurodegenerative disease. Seminars in cell & developmental biology. Elsevier; 2015.

  73. Pallotta MT, Rossini S, Suvieri C, Coletti A, Orabona C, Macchiarulo A, et al. Indoleamine 2, 3-dioxygenase 1 (IDO1): an up‐to‐date overview of an eclectic immunoregulatory enzyme. FEBS J. 2022;289(20):6099–118.

    Article  CAS  PubMed  Google Scholar 

  74. Feng W, Wang Y, Liu Z-Q, Zhang X, Han R, Miao Y-Z, et al. Microglia activation contributes to quinolinic acid-induced neuronal excitotoxicity through TNF-α. Apoptosis. 2017;22:696–709.

    Article  CAS  PubMed  Google Scholar 

  75. Rahman A, Ting K, Cullen KM, Braidy N, Brew BJ, Guillemin GJ. The excitotoxin quinolinic acid induces tau phosphorylation in human neurons. PLoS ONE. 2009;4(7):e6344.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Alachkar A, Agrawal S, Baboldashtian M, Nuseir K, Salazar J, Agrawal A. L-methionine enhances neuroinflammation and impairs neurogenesis: implication for Alzheimer’s disease. J Neuroimmunol. 2022;366:577843.

    Article  CAS  PubMed  Google Scholar 

  77. Zhao Y, Dong X, Chen B, Zhang Y, Meng S, Guo F, et al. Blood levels of circulating methionine components in Alzheimer’s disease and mild cognitive impairment: a systematic review and meta-analysis. Front Aging Neurosci. 2022;14:934070.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Miller AL. The methionine-homocysteine cycle and its effects on cognitive diseases.(Homocysteine & Cognitive). Altern Med Rev. 2003;8(1):7–20.

    PubMed  Google Scholar 

  79. Rath M, Müller I, Kropf P, Closs EI, Munder M. Metabolism via arginase or nitric oxide synthase: two competing arginine pathways in macrophages. Front Immunol. 2014;5:532.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Caldwell RW, Rodriguez PC, Toque HA, Narayanan SP, Caldwell RB. Arginase: a multifaceted enzyme important in health and disease. Physiol Rev. 2018;98(2):641–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Kan MJ, Lee JE, Wilson JG, Everhart AL, Brown CM, Hoofnagle AN, et al. Arginine deprivation and immune suppression in a mouse model of Alzheimer’s disease. J Neurosci. 2015;35(15):5969–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Ma C, Hunt JB, Kovalenko A, Liang H, Selenica M-LB, Orr MB, et al. Myeloid arginase 1 insufficiency exacerbates amyloid-β associated neurodegenerative pathways and glial signatures in a mouse model of Alzheimer’s disease: a targeted transcriptome analysis. Front Immunol. 2021;12:628156.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Urrutia P, Aguirre P, Esparza A, Tapia V, Mena NP, Arredondo M, et al. Inflammation alters the expression of DMT 1, FPN 1 and hepcidin, and it causes iron accumulation in central nervous system cells. J Neurochem. 2013;126(4):541–9.

    Article  CAS  PubMed  Google Scholar 

  84. Bishop GM, Dang TN, Dringen R, Robinson SR. Accumulation of non-transferrin-bound iron by neurons, astrocytes, and microglia. Neurotox Res. 2011;19:443–51.

    Article  CAS  PubMed  Google Scholar 

  85. Song N, Wang J, Jiang H, Xie J. Astroglial and microglial contributions to iron metabolism disturbance in Parkinson’s disease. Biochim et Biophys Acta (BBA)-Molecular Basis Disease. 2018;1864(3):967–73.

    Article  CAS  Google Scholar 

  86. Zeineh MM, Chen Y, Kitzler HH, Hammond R, Vogel H, Rutt BK. Activated iron-containing microglia in the human hippocampus identified by magnetic resonance imaging in Alzheimer disease. Neurobiol Aging. 2015;36(9):2483–500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. van Duijn S, Bulk M, van Duinen SG, Nabuurs RJ, van Buchem MA, van der Weerd L, et al. Cortical iron reflects severity of Alzheimer’s disease. J Alzheimers Dis. 2017;60(4):1533–45.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Holland R, McIntosh A, Finucane O, Mela V, Rubio-Araiz A, Timmons G, et al. Inflammatory microglia are glycolytic and iron retentive and typify the microglia in APP/PS1 mice. Brain Behav Immun. 2018;68:183–96.

    Article  CAS  PubMed  Google Scholar 

  89. McIntosh A, Mela V, Harty C, Minogue AM, Costello DA, Kerskens C, et al. Iron accumulation in microglia triggers a cascade of events that leads to altered metabolism and compromised function in APP/PS1 mice. Brain Pathol. 2019;29(5):606–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. The Multiple Sclerosis International Federation, Atlas of MS, 3rd Edition. (September 2020).

  91. Tortorella C, Bellacosa A, Paolicelli D, Fuiani A, Di Monte E, Simone IL, et al. Age-related gadolinium-enhancement of MRI brain lesions in multiple sclerosis. J Neurol Sci. 2005;239(1):95–9.

    Article  CAS  PubMed  Google Scholar 

  92. Giovannoni G, Silver N, Good C, Miller D, Thompson E. Immunological time-course of gadolinium-enhancing MRI lesions in patients with multiple sclerosis. Eur Neurol. 2000;44(4):222–8.

    Article  CAS  PubMed  Google Scholar 

  93. Brück W, Porada P, Poser S, Rieckmann P, Hanefeld F, Kretzschmarch HA, et al. Monocyte/macrophage differentiation in early multiple sclerosis lesions. Annals Neurology: Official J Am Neurol Association Child Neurol Soc. 1995;38(5):788–96.

    Article  Google Scholar 

  94. Guerrier T, Labalette M, Launay D, Lee-Chang C, Outteryck O, Lefèvre G, et al. Proinflammatory B-cell profile in the early phases of MS predicts an active disease. Neurology: Neuroimmunol Neuroinflammation. 2017;5(2):e431.

    Google Scholar 

  95. Murphy ÁC, Lalor SJ, Lynch MA, Mills KH. Infiltration of Th1 and Th17 cells and activation of microglia in the CNS during the course of experimental autoimmune encephalomyelitis. Brain Behav Immun. 2010;24(4):641–51.

    Article  CAS  PubMed  Google Scholar 

  96. Machado-Santos J, Saji E, Tröscher AR, Paunovic M, Liblau R, Gabriely G, et al. The compartmentalized inflammatory response in the multiple sclerosis brain is composed of tissue-resident CD8 + T lymphocytes and B cells. Brain. 2018;141(7):2066–82.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Lassmann H. Multiple sclerosis pathology. Cold Spring Harbor Perspect Med. 2018;8(3):a028936.

    Article  Google Scholar 

  98. Prineas JW, Parratt JD. Oligodendrocytes and the early multiple sclerosis lesion. Ann Neurol. 2012;72(1):18–31.

    Article  PubMed  Google Scholar 

  99. Wucherpfennig KW, Strominger JL. Molecular mimicry in T cell-mediated autoimmunity: viral peptides activate human T cell clones specific for myelin basic protein. Cell. 1995;80(5):695–705.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Maghzi A-H, Borazanci A, McGee J, Alexander JS, Gonzalez-Toledo E, Minagar A. Multiple sclerosis: pathophysiology, clinical features, diagnosis, and management. Neuroinflammation: Elsevier; 2011. pp. 1–23.

    Google Scholar 

  101. Hughes AN, Appel B. Microglia phagocytose myelin sheaths to modify developmental myelination. Nat Neurosci. 2020;23(9):1055–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Berglund R, Guerreiro-Cacais AO, Adzemovic MZ, Zeitelhofer M, Lund H, Ewing E, et al. Microglial autophagy–associated phagocytosis is essential for recovery from neuroinflammation. Sci Immunol. 2020;5(52):eabb5077.

    Article  CAS  PubMed  Google Scholar 

  103. Chéret C, Gervais A, Lelli A, Colin C, Amar L, Ravassard P, et al. Neurotoxic activation of microglia is promoted by a nox1-dependent NADPH oxidase. J Neurosci. 2008;28(46):12039–51.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Correale J. The role of microglial activation in disease progression. Multiple Scler J. 2014;20(10):1288–95.

    Article  CAS  Google Scholar 

  105. Broadwater L, Pandit A, Clements R, Azzam S, Vadnal J, Sulak M, et al. Analysis of the mitochondrial proteome in multiple sclerosis cortex. Biochim et Biophys Acta (BBA)-Molecular Basis Disease. 2011;1812(5):630–41.

    Article  CAS  Google Scholar 

  106. Talla V, Koilkonda R, Porciatti V, Chiodo V, Boye SL, Hauswirth WW, et al. Complex I subunit gene therapy with NDUFA6 ameliorates neurodegeneration in EAE. Investig Ophthalmol Vis Sci. 2015;56(2):1129–40.

    Article  CAS  Google Scholar 

  107. Regenold WT, Phatak P, Makley MJ, Stone RD, Kling MA. Cerebrospinal fluid evidence of increased extra-mitochondrial glucose metabolism implicates mitochondrial dysfunction in multiple sclerosis disease progression. J Neurol Sci. 2008;275(1–2):106–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Nijland PG, Molenaar RJ, van der Pol SM, van der Valk P, van Noorden CJ, de Vries HE, et al. Differential expression of glucose-metabolizing enzymes in multiple sclerosis lesions. Acta Neuropathol Commun. 2015;3:1–13.

    Article  Google Scholar 

  109. Keytsman C, Eijnde BO, Hansen D, Verboven K, Wens I. Elevated cardiovascular risk factors in multiple sclerosis. Multiple Scler Relat Disorders. 2017;17:220–3.

    Article  Google Scholar 

  110. Kolln J, Ren H-M, Da R-R, Zhang Y, Spillner E, Olek M, et al. Triosephosphate isomerase-and glyceraldehyde-3-phosphate dehydrogenase-reactive autoantibodies in the cerebrospinal fluid of patients with multiple sclerosis. J Immunol. 2006;177(8):5652–8.

    Article  CAS  PubMed  Google Scholar 

  111. Royds JA, Timperley WR, Taylor CB. Levels of enolase and other enzymes in the cerebrospinal fluid as indices of pathological change. J Neurol Neurosurg Psychiatry. 1981;44(12):1129–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Nijland PG, Michailidou I, Witte ME, Mizee MR, van der Pol SM, van Het Hof B, et al. Cellular distribution of glucose and monocarboxylate transporters in human brain white matter and multiple sclerosis lesions. Glia. 2014;62(7):1125–41.

    Article  PubMed  Google Scholar 

  113. Fantin VR, St-Pierre J, Leder P. Attenuation of LDH-A expression uncovers a link between glycolysis, mitochondrial physiology, and tumor maintenance. Cancer Cell. 2006;9(6):425–34.

    Article  CAS  PubMed  Google Scholar 

  114. Kaushik DK, Bhattacharya A, Mirzaei R, Rawji KS, Ahn Y, Rho JM, et al. Enhanced glycolytic metabolism supports transmigration of brain-infiltrating macrophages in multiple sclerosis. J Clin Investig. 2019;129(8):3277–92.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Gimeno-Bayón J, López‐López A, Rodríguez M, Mahy N. Glucose pathways adaptation supports acquisition of activated microglia phenotype. J Neurosci Res. 2014;92(6):723–31.

    Article  PubMed  Google Scholar 

  116. Bernier L-P, York EM, Kamyabi A, Choi HB, Weilinger NL, MacVicar BA. Microglial metabolic flexibility supports immune surveillance of the brain parenchyma. Nat Commun. 2020;11(1):1559.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Li Y, Lu B, Sheng L, Zhu Z, Sun H, Zhou Y, et al. Hexokinase 2-dependent hyperglycolysis driving microglial activation contributes to ischemic brain injury. J Neurochem. 2018;144(2):186–200.

    Article  CAS  PubMed  Google Scholar 

  118. Jha AK, Huang SC-C, Sergushichev A, Lampropoulou V, Ivanova Y, Loginicheva E, et al. Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization. Immunity. 2015;42(3):419–30.

    Article  CAS  PubMed  Google Scholar 

  119. Testa G, Staurenghi E, Zerbinati C, Gargiulo S, Iuliano L, Giaccone G, et al. Changes in brain oxysterols at different stages of Alzheimer’s disease: their involvement in neuroinflammation. Redox Biol. 2016;10:24–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Duc D, Vigne S, Pot C. Oxysterols in autoimmunity. Int J Mol Sci. 2019;20(18):4522.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. McComb M, Browne RW, Bhattacharya S, Bodziak ML, Jakimovski D, Weinstock-Guttman B, et al. The cholesterol autoxidation products, 7-ketocholesterol and 7β-hydroxycholesterol are associated with serum neurofilaments in multiple sclerosis. Multiple Scler Relat Disorders. 2021;50:102864.

    Article  CAS  Google Scholar 

  122. Diestel A, Aktas O, Hackel D, Häke I, Meier S, Raine CS, et al. Activation of microglial poly (ADP-ribose)-polymerase-1 by cholesterol breakdown products during neuroinflammation: a link between demyelination and neuronal damage. J Exp Med. 2003;198(11):1729–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Kauppinen TM, Swanson RA. Poly (ADP-ribose) polymerase-1 promotes microglial activation, proliferation, and matrix metalloproteinase-9-mediated neuron death. J Immunol. 2005;174(4):2288–96.

    Article  CAS  PubMed  Google Scholar 

  124. Bjørnevik K, Chitnis T, Ascherio A, Munger KL. Polyunsaturated fatty acids and the risk of multiple sclerosis. Multiple Scler J. 2017;23(14):1830–8.

    Article  Google Scholar 

  125. Hopperton KE, Trépanier M-O, Giuliano V, Bazinet RP. Brain omega-3 polyunsaturated fatty acids modulate microglia cell number and morphology in response to intracerebroventricular amyloid-β 1–40 in mice. J Neuroinflamm. 2016;13(1):1–18.

    Article  Google Scholar 

  126. Sun W, Wang Q, Zhang R, Zhang N. Ketogenic diet attenuates neuroinflammation and induces conversion of M1 microglia to M2 in an EAE model of multiple sclerosis by regulating the NF-κB/NLRP3 pathway and inhibiting HDAC3 and P2X7R activation. Food Funct. 2023;14(15):7247–69.

    Article  CAS  PubMed  Google Scholar 

  127. Piccio L, Buonsanti C, Cella M, Tassi I, Schmidt RE, Fenoglio C, et al. Identification of soluble TREM-2 in the cerebrospinal fluid and its association with multiple sclerosis and CNS inflammation. Brain. 2008;131(11):3081–91.

    Article  PubMed  PubMed Central  Google Scholar 

  128. Qu S, Hu S, Xu H, Wu Y, Ming S, Zhan X et al. TREM-2 drives development of multiple sclerosis by promoting pathogenic Th17 polarization. Neurosci Bull. 2023:1–18.

  129. Dong Y, D’Mello C, Pinsky W, Lozinski BM, Kaushik DK, Ghorbani S, et al. Oxidized phosphatidylcholines found in multiple sclerosis lesions mediate neurodegeneration and are neutralized by microglia. Nat Neurosci. 2021;24(4):489–503.

    Article  CAS  PubMed  Google Scholar 

  130. Takahashi K, Prinz M, Stagi M, Chechneva O, Neumann H. TREM2-transduced myeloid precursors mediate nervous tissue debris clearance and facilitate recovery in an animal model of multiple sclerosis. PLoS Med. 2007;4(4):e124.

    Article  PubMed  PubMed Central  Google Scholar 

  131. Piccio L, Buonsanti C, Mariani M, Cella M, Gilfillan S, Cross AH, et al. Blockade of TREM-2 exacerbates experimental autoimmune encephalomyelitis. Eur J Immunol. 2007;37(5):1290–301.

    Article  CAS  PubMed  Google Scholar 

  132. Negrotto L, Correale J. Amino acid catabolism in multiple sclerosis affects immune homeostasis. J Immunol. 2017;198(5):1900–9.

    Article  CAS  PubMed  Google Scholar 

  133. Kwidzinski E, Bunse J, Aktas O, Richter D, Mutlu L, Zipp F, et al. Indolamine 2, 3-dioxygenase is expressed in the CNS and down‐regulates autoimmune inflammation. FASEB J. 2005;19(10):1347–9.

    Article  CAS  PubMed  Google Scholar 

  134. Baydyuk M, Cha DS, Hu J, Yamazaki R, Miller EM, Smith VN et al. Tracking the evolution of CNS remyelinating lesion in mice with neutral red dye. Proceedings of the National Academy of Sciences. 2019;116(28):14290-9.

  135. Hu J, Melchor GS, Ladakis D, Reger J, Kim HW, Chamberlain KA, et al. Myeloid cell-associated aromatic amino acid metabolism facilitates CNS myelin regeneration. NPJ Regenerative Med. 2024;9(1):1.

    Article  Google Scholar 

  136. Fitzgerald KC, Smith MD, Kim S, Sotirchos ES, Kornberg MD, Douglas M et al. Multi-omic evaluation of metabolic alterations in multiple sclerosis identifies shifts in aromatic amino acid metabolism. Cell Rep Med. 2021;2(10).

  137. van der Poel M, Ulas T, Mizee MR, Hsiao C-C, Miedema SS, Adelia n, et al. Transcriptional profiling of human microglia reveals grey–white matter heterogeneity and multiple sclerosis-associated changes. Nat Commun. 2019;10(1):1139.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Mehta V, Pei W, Yang G, Li S, Swamy E, Boster A, et al. Iron is a sensitive biomarker for inflammation in multiple sclerosis lesions. PLoS ONE. 2013;8(3):e57573.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Khalil M, Teunissen C, Langkammer C. Iron and neurodegeneration in multiple sclerosis. Multiple sclerosis international. 2011;2011.

  140. Proto JD, Zhang M, Ryan S, Yao X, Huang Y, Chang Y-C et al. Disrupted microglial iron homeostasis in progressive multiple sclerosis. bioRxiv. 2021:2021.05. 09.443127.

  141. Lee DW, Andersen JK. Role of HIF-1 in iron regulation: potential therapeutic strategy for neurodegenerative disorders. Curr Mol Med. 2006;6(8):883–93.

    Article  CAS  PubMed  Google Scholar 

  142. Rissanen E, Tuisku J, Rokka J, Paavilainen T, Parkkola R, Rinne JO, et al. In vivo detection of diffuse inflammation in secondary progressive multiple sclerosis using PET imaging and the radioligand 11 C-PK11195. J Nucl Med. 2014;55(6):939–44.

    Article  CAS  PubMed  Google Scholar 

  143. Marras C, Beck J, Bower J, Roberts E, Ritz B, Ross G, et al. Prevalence of Parkinson’s disease across North America. NPJ Parkinson’s Disease. 2018;4(1):21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Balestrino R, Schapira A. Parkinson disease. Eur J Neurol. 2020;27(1):27–42.

    Article  CAS  PubMed  Google Scholar 

  145. Simon DK, Tanner CM, Brundin P. Parkinson disease epidemiology, pathology, genetics, and pathophysiology. Clin Geriatr Med. 2020;36(1):1–12.

    Article  PubMed  Google Scholar 

  146. Zhu B, Yin D, Zhao H, Zhang L, editors. The immunology of Parkinson’s disease. Seminars in Immunopathology. Springer; 2022.

  147. Lee H-J, Suk J-E, Bae E-J, Lee S-J. Clearance and deposition of extracellular α-synuclein aggregates in microglia. Biochem Biophys Res Commun. 2008;372(3):423–8.

    Article  CAS  PubMed  Google Scholar 

  148. Xia Y, Zhang G, Han C, Ma K, Guo X, Wan F, et al. Microglia as modulators of exosomal alpha-synuclein transmission. Cell Death Dis. 2019;10(3):174.

    Article  PubMed  PubMed Central  Google Scholar 

  149. Emmanouilidou E, Melachroinou K, Roumeliotis T, Garbis SD, Ntzouni M, Margaritis LH, et al. Cell-produced α-synuclein is secreted in a calcium-dependent manner by exosomes and impacts neuronal survival. J Neurosci. 2010;30(20):6838–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Danzer KM, Kranich LR, Ruf WP, Cagsal-Getkin O, Winslow AR, Zhu L, et al. Exosomal cell-to-cell transmission of alpha synuclein oligomers. Mol Neurodegeneration. 2012;7:1–18.

    Article  Google Scholar 

  151. Bliederhaeuser C, Grozdanov V, Speidel A, Zondler L, Ruf WP, Bayer H, et al. Age-dependent defects of alpha-synuclein oligomer uptake in microglia and monocytes. Acta Neuropathol. 2016;131:379–91.

    Article  CAS  PubMed  Google Scholar 

  152. Zhang W, Phillips K, Wielgus AR, Liu J, Albertini A, Zucca FA, et al. Neuromelanin activates microglia and induces degeneration of dopaminergic neurons: implications for progression of Parkinson’s disease. Neurotox Res. 2011;19:63–72.

    Article  PubMed  Google Scholar 

  153. Rueda-Carrasco J, Sokolova D, Lee SE, Childs T, Jurčáková N, Crowley G, et al. Microglia‐synapse engulfment via PtdSer‐TREM2 ameliorates neuronal hyperactivity in Alzheimer’s disease models. EMBO J. 2023;42(19):e113246.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Badanjak K, Fixemer S, Smajić S, Skupin A, Grünewald A. The contribution of microglia to neuroinflammation in Parkinson’s disease. Int J Mol Sci. 2021;22(9):4676.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Calsolaro V, Edison P. Neuroinflammation in Alzheimer’s disease: current evidence and future directions. Alzheimer’s Dement. 2016;12(6):719–32.

    Article  Google Scholar 

  156. Dunn L, Allen GF, Mamais A, Ling H, Li A, Duberley KE, et al. Dysregulation of glucose metabolism is an early event in sporadic Parkinson’s disease. Neurobiol Aging. 2014;35(5):1111–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Edison P, Ahmed I, Fan Z, Hinz R, Gelosa G, Ray Chaudhuri K, et al. Microglia, amyloid, and glucose metabolism in Parkinson’s disease with and without dementia. Neuropsychopharmacology. 2013;38(6):938–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Tai YF, Pavese N, Gerhard A, Tabrizi SJ, Barker RA, Brooks DJ, et al. Imaging microglial activation in Huntington’s disease. Brain Res Bull. 2007;72(2–3):148–51.

    Article  CAS  PubMed  Google Scholar 

  159. Iannaccone S, Cerami C, Alessio M, Garibotto V, Panzacchi A, Olivieri S, et al. In vivo microglia activation in very early dementia with Lewy bodies, comparison with Parkinson’s disease. Parkinsonism Relat Disord. 2013;19(1):47–52.

    Article  CAS  PubMed  Google Scholar 

  160. Qiao H, He X, Zhang Q, Yuan H, Wang D, Li L, et al. Alpha-synuclein induces microglial migration via PKM2-dependent glycolysis. Int J Biol Macromol. 2019;129:601–7.

    Article  CAS  PubMed  Google Scholar 

  161. Stefanis L. α-Synuclein in Parkinson’s disease. Cold Spring Harbor Perspect Med. 2012;2(2):a009399.

    Article  Google Scholar 

  162. Rodriguez-Araujo G, Nakagami H, Hayashi H, Mori M, Shiuchi T, Minokoshi Y, et al. Alpha-synuclein elicits glucose uptake and utilization in adipocytes through the Gab1/PI3K/Akt transduction pathway. Cell Mol Life Sci. 2013;70:1123–33.

    Article  CAS  PubMed  Google Scholar 

  163. Lu J, Wang C, Cheng X, Wang R, Yan X, He P, et al. A breakdown in microglial metabolic reprogramming causes internalization dysfunction of α-synuclein in a mouse model of Parkinson’s disease. J Neuroinflamm. 2022;19(1):1–21.

    Article  Google Scholar 

  164. Qiao H, He X, Zhang Q, Zhang N, Li L, Hui Y, et al. Α-synuclein induces microglial cell migration through stimulating HIF‐1α accumulation. J Neurosci Res. 2017;95(9):1809–17.

    Article  CAS  PubMed  Google Scholar 

  165. Nakai D, Yuasa S, Takahashi M, Shimizu T, Asaumi S, Isono K, et al. Mouse homologue of coq7/clk-1, longevity gene in Caenorhabditis elegans, is essential for coenzyme Q synthesis, maintenance of mitochondrial integrity, and neurogenesis. Biochem Biophys Res Commun. 2001;289(2):463–71.

    Article  CAS  PubMed  Google Scholar 

  166. Gu R, Zhang F, Chen G, Han C, Liu J, Ren Z, et al. Clk1 deficiency promotes neuroinflammation and subsequent dopaminergic cell death through regulation of microglial metabolic reprogramming. Brain Behav Immun. 2017;60:206–19.

    Article  CAS  PubMed  Google Scholar 

  167. Joshi L, Plastira I, Bernhart E, Reicher H, Koyani CN, Madl T, et al. Lysophosphatidic acid induces aerobic glycolysis, lipogenesis, and increased amino acid uptake in BV-2 microglia. Int J Mol Sci. 2021;22(4):1968.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Panicker N, Sarkar S, Harischandra DS, Neal M, Kam T-I, Jin H, et al. Fyn kinase regulates misfolded α-synuclein uptake and NLRP3 inflammasome activation in microglia. J Exp Med. 2019;216(6):1411.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Arab HH, Safar MM, Shahin NN. Targeting ROS-dependent AKT/GSK-3β/NF-κB and DJ-1/Nrf2 pathways by dapagliflozin attenuates neuronal injury and motor dysfunction in rotenone-induced Parkinson’s disease rat model. ACS Chem Neurosci. 2021;12(4):689–703.

    Article  CAS  PubMed  Google Scholar 

  170. Zhou Q, Zhang Y, Lu L, Zhang H, Zhao C, Pu Y, et al. Copper induces microglia-mediated neuroinflammation through ROS/NF-κB pathway and mitophagy disorder. Food Chem Toxicol. 2022;168:113369.

    Article  CAS  PubMed  Google Scholar 

  171. Takeuchi H, Jin S, Wang J, Zhang G, Kawanokuchi J, Kuno R, et al. Tumor necrosis factor-α induces neurotoxicity via glutamate release from hemichannels of activated microglia in an autocrine manner. J Biol Chem. 2006;281(30):21362–8.

    Article  CAS  PubMed  Google Scholar 

  172. Reynolds AD, Kadiu I, Garg SK, Glanzer JG, Nordgren T, Ciborowski P, et al. Nitrated alpha-synuclein and microglial neuroregulatory activities. J Neuroimmune Pharmacol. 2008;3:59–74.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Bak LK, Schousboe A, Waagepetersen HS. The glutamate/GABA-glutamine cycle: aspects of transport, neurotransmitter homeostasis and ammonia transfer. J Neurochem. 2006;98(3):641–53.

    Article  CAS  PubMed  Google Scholar 

  174. Lind-Holm Mogensen F, Sousa C, Ameli C, Badanjak K, Pereira SL, Muller A, et al. PARK7/DJ-1 deficiency impairs microglial activation in response to LPS-induced inflammation. J Neuroinflamm. 2024;21(1):174.

    Article  CAS  Google Scholar 

  175. Brekk OR, Honey JR, Lee S, Hallett PJ, Isacson O. Cell type-specific lipid storage changes in Parkinson’s disease patient brains are recapitulated by experimental glycolipid disturbance. Proceedings of the National Academy of Sciences. 2020;117(44):27646-54.

  176. Marschallinger J, Iram T, Zardeneta M, Lee SE, Lehallier B, Haney MS, et al. Lipid-droplet-accumulating microglia represent a dysfunctional and proinflammatory state in the aging brain. Nat Neurosci. 2020;23(2):194–208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Loving BA, Tang M, Neal MC, Gorkhali S, Murphy R, Eckel RH, et al. Lipoprotein lipase regulates microglial lipid droplet accumulation. Cells. 2021;10(2):198.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Illarioshkin S, Klyushnikov S, Vigont V, Seliverstov YA, Kaznacheyeva E. Molecular pathogenesis in Huntington’s disease. Biochem (Moscow). 2018;83:1030–9.

    Article  CAS  Google Scholar 

  179. Yang H-M, Yang S, Huang S-S, Tang B-S, Guo J-F. Microglial activation in the pathogenesis of Huntington’s disease. Front Aging Neurosci. 2017;9:193.

    Article  PubMed  PubMed Central  Google Scholar 

  180. Palpagama TH, Waldvogel HJ, Faull RL, Kwakowsky A. The role of microglia and astrocytes in Huntington’s disease. Front Mol Neurosci. 2019;12:258.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Khoshnan A, Ko J, Watkin EE, Paige LA, Reinhart PH, Patterson PH. Activation of the IκB kinase complex and nuclear factor-κB contributes to mutant huntingtin neurotoxicity. J Neurosci. 2004;24(37):7999–8008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Khoshnan A, Patterson PH. The role of IκB kinase complex in the neurobiology of Huntington’s disease. Neurobiol Dis. 2011;43(2):305–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Schwarcz R, Guidetti P, Sathyasaikumar KV, Muchowski PJ. Of mice, rats and men: revisiting the quinolinic acid hypothesis of Huntington’s disease. Prog Neurobiol. 2010;90(2):230–45.

    Article  CAS  PubMed  Google Scholar 

  184. Palazuelos J, Aguado T, Pazos MR, Julien B, Carrasco C, Resel E, et al. Microglial CB2 cannabinoid receptors are neuroprotective in Huntington’s disease excitotoxicity. Brain. 2009;132(11):3152–64.

    Article  PubMed  Google Scholar 

  185. Reijonen S, Kukkonen JP, Hyrskyluoto A, Kivinen J, Kairisalo M, Takei N, et al. Downregulation of NF-κB signaling by mutant huntingtin proteins induces oxidative stress and cell death. Cell Mol Life Sci. 2010;67:1929–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Bečanović K, Nørremølle A, Neal SJ, Kay C, Collins JA, Arenillas D, et al. A SNP in the HTT promoter alters NF-κB binding and is a bidirectional genetic modifier of Huntington disease. Nat Neurosci. 2015;18(6):807–16.

    Article  PubMed  Google Scholar 

  187. Marcora E, Kennedy MB. The Huntington’s disease mutation impairs Huntingtin’s role in the transport of NF-κB from the synapse to the nucleus. Hum Mol Genet. 2010;19(22):4373–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Träger U, Andre R, Lahiri N, Magnusson-Lind A, Weiss A, Grueninger S, et al. HTT-lowering reverses Huntington’s disease immune dysfunction caused by NFκB pathway dysregulation. Brain. 2014;137(3):819–33.

    Article  PubMed  PubMed Central  Google Scholar 

  189. Reddy V, Grogan D, Ahluwalia M, Salles ÉL, Ahluwalia P, Khodadadi H, et al. Targeting the endocannabinoid system: a predictive, preventive, and personalized medicine-directed approach to the management of brain pathologies. EPMA J. 2020;11:217–50.

    Article  PubMed  PubMed Central  Google Scholar 

  190. Wang Q, Zhao Y, Sun M, Liu S, Li B, Zhang L, et al. 2-Deoxy-d-glucose attenuates sevoflurane-induced neuroinflammation through nuclear factor-kappa B pathway in vitro. Toxicol in Vitro. 2014;28(7):1183–9.

    Article  CAS  PubMed  Google Scholar 

  191. Badawy AA. Kynurenine pathway of tryptophan metabolism: regulatory and functional aspects. Int J Tryptophan Res. 2017;10:1178646917691938.

    Article  PubMed  PubMed Central  Google Scholar 

  192. Giorgini F, Moller T, Kwan W, Zwilling D, Wacker JL, Hong S, et al. Histone deacetylase inhibition modulates kynurenine pathway activation in yeast, microglia, and mice expressing a mutant huntingtin fragment. J Biol Chem. 2008;283(12):7390–400.

    Article  CAS  PubMed  Google Scholar 

  193. Ryu JK, Kim SU, McLarnon JG. Blockade of quinolinic acid-induced neurotoxicity by pyruvate is associated with inhibition of glial activation in a model of Huntington’s disease. Exp Neurol. 2004;187(1):150–9.

    Article  CAS  PubMed  Google Scholar 

  194. Tan Z, Xie N, Cui H, Moellering DR, Abraham E, Thannickal VJ, et al. Pyruvate dehydrogenase kinase 1 participates in macrophage polarization via regulating glucose metabolism. J Immunol. 2015;194(12):6082–9.

    Article  CAS  PubMed  Google Scholar 

  195. Van Bergen JM, Hua J, Unschuld PG, Lim IAL, Jones CK, Margolis RL, et al. Quantitative susceptibility mapping suggests altered brain iron in Premanifest Huntington disease. Am J Neuroradiol. 2016;37(5):789–96.

    Article  PubMed  PubMed Central  Google Scholar 

  196. Bartzokis G, Cummings J, Perlman S, Hance DB, Mintz J. Increased basal ganglia iron levels in Huntington disease. Arch Neurol. 1999;56(5):569–74.

    Article  CAS  PubMed  Google Scholar 

  197. Simmons DA, Casale M, Alcon B, Pham N, Narayan N, Lynch G. Ferritin accumulation in dystrophic microglia is an early event in the development of Huntington’s disease. Glia. 2007;55(10):1074–84.

    Article  PubMed  Google Scholar 

  198. González-Guevara E, Cárdenas G, Pérez‐Severiano F, Martínez‐Lazcano JC. Dysregulated brain cholesterol metabolism is linked to neuroinflammation in Huntington’s disease. Mov Disord. 2020;35(7):1113–27.

    Article  PubMed  Google Scholar 

  199. Brown RH, Al-Chalabi A. Amyotrophic lateral sclerosis. N Engl J Med. 2017;377(2):162–72.

    Article  CAS  PubMed  Google Scholar 

  200. Rothstein JD. Current hypotheses for the underlying biology of amyotrophic lateral sclerosis. Ann Neurol. 2009;65(S1):S3–9.

    Article  CAS  PubMed  Google Scholar 

  201. Beers DR, Henkel JS, Zhao W, Wang J, Appel SH. CD4 + T cells support glial neuroprotection, slow disease progression, and modify glial morphology in an animal model of inherited ALS. Proceedings of the National Academy of Sciences. 2008;105(40):15558-63.

  202. Philips T, Robberecht W. Neuroinflammation in amyotrophic lateral sclerosis: role of glial activation in motor neuron disease. Lancet Neurol. 2011;10(3):253–63.

    Article  CAS  PubMed  Google Scholar 

  203. Funalot B, Desport J-C, Sturtz F, Camu W, Couratier P. High metabolic level in patients with familial amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2009;10(2):113–7.

    Article  CAS  PubMed  Google Scholar 

  204. Vandoorne T, De Bock K, Van Den Bosch L. Energy metabolism in ALS: an underappreciated opportunity? Acta Neuropathol. 2018;135:489–509.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Tefera TW, Borges K. Metabolic dysfunctions in amyotrophic lateral sclerosis pathogenesis and potential metabolic treatments. Front NeuroSci. 2017;10:611.

    Article  PubMed  PubMed Central  Google Scholar 

  206. Vallée A, Lecarpentier Y, Guillevin R, Vallée J-N. Aerobic glycolysis in amyotrophic lateral sclerosis and Huntington’s disease. Rev Neurosci. 2018;29(5):547–55.

    Article  PubMed  Google Scholar 

  207. Wiedemann FR, Manfredi G, Mawrin C, Beal MF, Schon EA. Mitochondrial DNA and respiratory chain function in spinal cords of ALS patients. J Neurochem. 2002;80(4):616–25.

    Article  CAS  PubMed  Google Scholar 

  208. Borthwick GM, Johnson MA, Ince PG, Shaw PJ, Turnbull DM. Mitochondrial enzyme activity in amyotrophic lateral sclerosis: implications for the role of mitochondria in neuronal cell death. Annals Neurology: Official J Am Neurol Association Child Neurol Soc. 1999;46(5):787–90.

    Article  CAS  Google Scholar 

  209. Carrì MT, D’Ambrosi N, Cozzolino M. Pathways to mitochondrial dysfunction in ALS pathogenesis. Biochem Biophys Res Commun. 2017;483(4):1187–93.

    Article  PubMed  Google Scholar 

  210. Sunyach C, Michaud M, Arnoux T, Bernard-Marissal N, Aebischer J, Latyszenok V, et al. Olesoxime delays muscle denervation, astrogliosis, microglial activation and motoneuron death in an ALS mouse model. Neuropharmacology. 2012;62(7):2346–53.

    Article  CAS  PubMed  Google Scholar 

  211. Niida-Kawaguchi M, Kakita A, Noguchi N, Kazama M, Masui K, Kato Y, et al. Soluble iron accumulation induces microglial glutamate release in the spinal cord of sporadic amyotrophic lateral sclerosis. Neuropathology. 2020;40(2):152–66.

    Article  CAS  PubMed  Google Scholar 

  212. Milanese M, Giribaldi F, Melone M, Bonifacino T, Musante I, Carminati E, et al. Knocking down metabotropic glutamate receptor 1 improves survival and disease progression in the SOD1G93A mouse model of amyotrophic lateral sclerosis. Neurobiol Dis. 2014;64:48–59.

    Article  CAS  PubMed  Google Scholar 

  213. Parone PA, Da Cruz S, Han JS, McAlonis-Downes M, Vetto AP, Lee SK, et al. Enhancing mitochondrial calcium buffering capacity reduces aggregation of misfolded SOD1 and motor neuron cell death without extending survival in mouse models of inherited amyotrophic lateral sclerosis. J Neurosci. 2013;33(11):4657–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Bonifacino T, Cattaneo L, Gallia E, Puliti A, Melone M, Provenzano F, et al. In-vivo effects of knocking-down metabotropic glutamate receptor 5 in the SOD1G93A mouse model of amyotrophic lateral sclerosis. Neuropharmacology. 2017;123:433–45.

    Article  CAS  PubMed  Google Scholar 

  215. Hu Y, Cao K, Wang F, Wu W, Mai W, Qiu L, et al. Dual roles of hexokinase 2 in shaping microglial function by gating glycolytic flux and mitochondrial activity. Nat Metabolism. 2022;4(12):1756–74.

    Article  CAS  Google Scholar 

  216. Haney MS, Pálovics R, Munson CN, Long C, Johansson PK, Yip O et al. APOE4/4 is linked to damaging lipid droplets in Alzheimer’s disease microglia. Nature. 2024:1–8.

  217. Hopperton KE, Trépanier M-O, Giuliano V, Bazinet RP. Brain omega-3 polyunsaturated fatty acids modulate microglia cell number and morphology in response to intracerebroventricular amyloid-β 1–40 in mice. J Neuroinflamm. 2016;13:1–18.

    Article  Google Scholar 

  218. Leng L, Yuan Z, Pan R, Su X, Wang H, Xue J, et al. Microglial hexokinase 2 deficiency increases ATP generation through lipid metabolism leading to β-amyloid clearance. Nat Metabolism. 2022;4(10):1287–305.

    Article  CAS  Google Scholar 

  219. Gonzalez-Gil A, Porell RN, Fernandes SM, Maenpaa E, Li TA, Li T et al. Human brain sialoglycan ligand for CD33, a microglial inhibitory siglec implicated in Alzheimer’s disease. J Biol Chem. 2022;298(6).

  220. Lin Y-T, Seo J, Gao F, Feldman HM, Wen H-L, Penney J, et al. APOE4 causes widespread molecular and cellular alterations associated with Alzheimer’s disease phenotypes in human iPSC-derived brain cell types. Neuron. 2018;98(6):1141–54. e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Victor MB, Leary N, Luna X, Meharena HS, Scannail AN, Bozzelli PL, et al. Lipid accumulation induced by APOE4 impairs microglial surveillance of neuronal-network activity. Cell Stem Cell. 2022;29(8):1197–212. e8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Liu C-C, Wang N, Chen Y, Inoue Y, Shue F, Ren Y, et al. Cell-autonomous effects of APOE4 in restricting microglial response in brain homeostasis and Alzheimer’s disease. Nat Immunol. 2023;24(11):1854–66.

    Article  CAS  PubMed  Google Scholar 

  223. Lee S, Devanney NA, Golden LR, Smith CT, Schwartz JL, Walsh AE et al. APOE modulates microglial immunometabolism in response to age, amyloid pathology, and inflammatory challenge. Cell Rep. 2023;42(3).

  224. Zrzavy T, Hametner S, Wimmer I, Butovsky O, Weiner HL, Lassmann H. Loss of ‘homeostatic’microglia and patterns of their activation in active multiple sclerosis. Brain. 2017;140(7):1900–13.

    Article  PubMed  PubMed Central  Google Scholar 

  225. Shijie J, Takeuchi H, Yawata I, Harada Y, Sonobe Y, Doi Y, et al. Blockade of glutamate release from microglia attenuates experimental autoimmune encephalomyelitis in mice. Tohoku J Exp Med. 2009;217(2):87–92.

    Article  CAS  PubMed  Google Scholar 

  226. Meng F, Yu W, Duan W, Wang T, Liu Y. Dexmedetomidine attenuates LPS-mediated BV2 microglia cells inflammation via inhibition of glycolysis. Fundam Clin Pharmacol. 2020;34(3):313–20.

    Article  CAS  PubMed  Google Scholar 

  227. Sradhanjali S, Reddy MM. Inhibition of pyruvate dehydrogenase kinase as a therapeutic strategy against Cancer. Curr Top Med Chem. 2018;18(6):444–53.

    Article  CAS  PubMed  Google Scholar 

  228. Ramanan S, Kooshki M, Zhao W, Hsu FC, Riddle DR, Robbins ME. The PPARalpha agonist fenofibrate preserves hippocampal neurogenesis and inhibits microglial activation after whole-brain irradiation. Int J Radiat Oncol Biol Phys. 2009;75(3):870–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Bernardo A, Minghetti L. PPAR-γ Agonists as Regulators of Microglial Activation and Brain Inflammation. Current Pharmaceutical Design. 2006;12(1):93–109.

  230. Carta AR, Pisanu A. Modulating microglia activity with PPAR-γ agonists: a promising therapy for Parkinson’s disease? Neurotox Res. 2013;23(2):112–23.

    Article  CAS  PubMed  Google Scholar 

  231. Stratoulias V, Venero JL, Tremblay MÈ, Joseph B. Microglial subtypes: diversity within the microglial community. EMBO J. 2019;38(17):e101997.

    Article  PubMed  PubMed Central  Google Scholar 

  232. Vilalta A, Brown GC. Deoxyglucose prevents neurodegeneration in culture by eliminating microglia. J Neuroinflamm. 2014;11:1–10.

    Article  Google Scholar 

  233. He D, Xu H, Zhang H, Tang R, Lan Y, Xing R, et al. Disruption of the IL-33-ST2-AKT signaling axis impairs neurodevelopment by inhibiting microglial metabolic adaptation and phagocytic function. Immunity. 2022;55(1):159–73. e9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Lauro C, Limatola C. Metabolic reprograming of microglia in the regulation of the innate inflammatory response. Front Immunol. 2020;11:523362.

    Article  Google Scholar 

  235. Mikitsh JL, Chacko A-M. Pathways for small molecule delivery to the central nervous system across the blood-brain barrier. Perspect Med Chem. 2014;6:PMC.

    Google Scholar 

Download references

Acknowledgements

Figures were created via BioRender.com.

Funding

This work was supported by an Exploration – Hypothesis Development grant from the Congressionally Directed Multiple Sclerosis Program of the USA Department of Defense under Award No. MS220110 (DKK) and MS Canada (DKK). Opinions, interpretations, conclusions, and recommendations are those of the authors and are not necessarily endorsed by the Department of Defense.

Author information

Authors and Affiliations

Authors

Contributions

DK, MS and AD conceptualized the work. MS and AD wrote the original draft. DK supervised and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Deepak Kumar Kaushik.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sadeghdoust, M., Das, A. & Kaushik, D.K. Fueling neurodegeneration: metabolic insights into microglia functions. J Neuroinflammation 21, 300 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12974-024-03296-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12974-024-03296-0

Keywords