
Lipid metabolism in microglia: Emerging mechanisms and therapeutic opportunities for neurodegenerative diseases (Review)
- Authors:
- Published online on: July 8, 2025 https://doi.org/10.3892/ijmm.2025.5580
- Article Number: 139
-
Copyright: © Sun et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Neurodegenerative diseases (NDDs) represent a group of disorders characterized by progressive degeneration and neuronal death within the nervous system. With accelerating global population aging, NDDs have emerged as a critical public health concern, imposing substantial burdens on individual health outcomes and societal healthcare resources (1). Alzheimer's disease (AD), Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS) constitute the most prevalent and clinically severe representatives within this category (2). AD manifests as progressive cognitive decline accompanied by β-amyloid aggregation and neurofibrillary tangles (3). PD primarily presents with motor symptoms including tremor, rigidity and bradykinesia, with degeneration of dopaminergic neurons in the substantia nigra representing its core pathological hallmark (4). ALS features progressive muscular weakness and atrophy resulting from extensive upper and lower motor neuron degeneration (5). Despite distinct clinical presentations, these disorders share common pathological mechanisms involving neuronal damage, apoptosis and neuroinflammation, with disease pathogenesis intricately linked to gene-environment interactions (6,7).
Microglia, the resident immune cells of the central nervous system (CNS), play pivotal roles in maintaining neural homeostasis and responding to pathological perturbations (8,9). Under physiological conditions, quiescent microglia continuously surveil the neural microenvironment through dynamic process extension, participating in neurodevelopmental processes and synaptic pruning (10). However, in neurodegenerative contexts-particularly in AD, PD and ALS-microglial functions undergo significant alterations (11-13). Activated microglia proliferate and secrete cytokines, chemokines and immune mediators, initiating neuroinflammatory cascades that exacerbate neuronal damage (14). Studies demonstrate that chronic microglial activation correlates with sustained neuroinflammation, potentially accelerating neurodegeneration through oxidative stress induction, immune dysregulation and apoptotic pathway activation (15,16).
Lipid metabolism maintains fundamental importance in preserving cellular structural and functional integrity (17). Lipids not only serve as fundamental structural components of cellular membranes but also participate in signal transduction, energy storage and various cellular regulatory processes. Of note, dysregulated lipid metabolism in microglia may critically influence neurodegenerative pathogenesis through neuroinflammatory modulation (12,18,19). Research indicates that fatty acid metabolites, particularly oxidized lipids, can regulate microglial immune responses by activating specific receptor pathways (20,21). Accumulation of lipid peroxidation products, especially oxidized long-chain fatty acids, induces microglial hyperactivation and pro-inflammatory cytokine release, thereby initiating or amplifying neural damage (18). Furthermore, lipid metabolic disturbances may contribute to neuronal membrane deterioration and functional decline, suggesting their potential utility as biomarkers and therapeutic targets in NDDs.
Microglial activation in NDDs
Pattern recognition and phenotypic transition of microglia
As the principal immune effector cells in the CNS, microglial activation constitutes a central event in neurodegenerative pathogenesis. In NDDs, microglia detect pathological signals through pattern recognition receptors (PRRs), including damage-associated molecular patterns (DAMPs) such as amyloid-β (Aβ) aggregates, hyperphosphorylated Tau proteins, neuronal debris and mitochondrial DNA (mtDNA) in AD (22,23). Disease-specific DAMP activation mechanisms also operate across other NDDs: For instance, α-synuclein (α-syn) oligomers in PD activate the NF-κB signaling pathway via Toll-like receptor (TLR)2/4 receptors (24), while myelin fragments in multiple sclerosis (MS)-which is primarily an autoimmune demyelinating disease-trigger inflammation through TLR4/MYD88 innate immune signal transduction adaptor (MyD88) pathways (25,26). These endogenous signals induce microglial transition from resting (M0) to activated states, manifesting as somatic hypertrophy, process retraction and enhanced motility (27,28). Notably, microglial activation exhibits high heterogeneity, with phenotypes dynamically regulated by local microenvironmental cues. For instance, Aβ oligomers bind CD36/TLR4/TLR6 receptor complexes to activate MyD88-dependent signaling, driving nuclear translocation of NF-κB and interferon regulatory factors, thereby initiating transcription of pro-inflammatory cytokines (e.g., IL-1β, TNF-α) and chemokines [e.g., C-C motif chemokine ligand (CCL)2, CXCL10] (29,30). Similarly, α-syn in PD activates the NLR family pyrin domain containing 3 (NLRP3) inflammasome via caspase-1-dependent pyroptosis, promoting gasdermin D cleavage (31), while TAR DNA binding protein-43 aggregates in ALS trigger calcium influx-dependent NLRP3 activation through P2X7 receptors (32,33). These mechanisms amplify inflammatory signaling and facilitate NLRP3 inflammasome assembly (34,35).
Aberrant microglial endocytosis critically modulates neurodegenerative microenvironments. In AD pathology, abnormally modified Tau proteins regulate microglial activation: Hyperphosphorylated Tau enters microglia via CX3CR1 receptor-mediated endocytosis, activating Syk tyrosine kinase signaling to induce phagolysosomal dysfunction and pro-inflammatory mediator release (36,37). This receptor-mediated endocytic mechanism demonstrates cross-disease conservation: In Huntington's disease (HD), mutant huntingtin upregulates complement C1q-mediated synaptic tagging of PSD-95 sites, activating CR3-dependent synaptic pruning (38-40), while progranulin deficiency in frontotemporal dementia exacerbates Tau propagation through lysosomal dysfunction (41). In addition, ATP released from injured neurons activates P2X7 purinergic receptors to induce calcium oscillations, enhancing NLRP3 inflammasome activity and caspase-1-dependent IL-1β maturation (35,42). In traumatic brain injury and neuroinflammation, triggering receptor expressed on myeloid cells 1 (TREM-1) receptors mediate microglia-neutrophil crosstalk via SYK signaling, amplifying dopaminergic neuron degeneration in the substantia nigra (43). Such multi-signal synergy sustains microglial activation, establishing self-reinforcing inflammatory loops. Importantly, aging-associated microglial senescence elevates activation thresholds: TREM2 downregulation in aged individuals reduces Aβ clearance efficiency, while senescence-associated secretory phenotype factors (e.g., IL-6, MMP-9) impair homeostatic maintenance (44,45).
Microglial activation exhibits stage-dependent characteristics in neurodegenerative progression. During early AD, localized Aβ deposits recruit microglia via chemokine gradients to form barrier-like structures constraining plaque spread, with microglia predominantly exhibiting M2-like anti-inflammatory phenotypes that secrete insulin-degrading enzyme and apolipoprotein E (APOE) to facilitate Aβ clearance (46,47). Similar spatial heterogeneity occurs in MS lesions: Peri-plaque microglia overexpress APOE and secreted phosphoprotein 1 (SPP1), displaying enhanced phagocytosis but impaired repair capacity (48,49), while C1q complement inhibition rescues aberrant synaptic pruning (50,51). However, chronic inflammatory microenvironments drive phenotypic shifts toward M1-like proinflammatory states as pathology advances. Transcriptomic studies reveal that disease-associated microglia in AD brains upregulate APOE, TREM2 and SPP1 while downregulating phagocytic pathways [e.g., liver X receptor (LXR)/retinoid X receptor (RXR) signaling], suggesting functional transition from protective clearance to pathological promotion (44). In HD, this phenotypic shift involves cyclic GMP-AMP synthase (cGAS)-stimulator of interferon response cGAMP interactor (STING) pathway activation: Mutant Huntingtin (mHTT) induces mtDNA leakage, triggering chronic neuroinflammation via type I interferon responses (52-54). Such transitions correlate with mitochondrial dysfunction: Aβ-induced loss of mitochondrial membrane potential reduces ATP synthesis, while leaked mtDNA activates cGAS-STING signaling to exacerbate type I interferon production and neuroinflammation (55,56).
Balancing act: Pro-inflammatory injury vs. anti-inflammatory repair
Microglia exhibit dual paradoxical functions in NDDs: Their pro-inflammatory (M1) phenotype exacerbates neuronal damage through cytotoxic mediator release, while the anti-inflammatory (M2) phenotype exerts neuroprotection via pathological protein clearance and neurotrophic factor secretion (as shown in Fig. 1). This functional dichotomy is dynamically regulated by cytokine networks, disease stages and aging status. During early AD, M2-polarized microglia clear soluble Aβ through TREM2-dependent phagocytosis and secrete brain-derived neurotrophic factor and TGF-β to enhance synaptic plasticity (46,47,57). Furthermore, IL-4 stimulation promotes M2 polarization via STAT6 signaling-mediated upregulation of arginase-1 (Arg1) and chitinase-like molecule 1, enhancing Aβ degradation capacity while suppressing neuroinflammation (58,59).
However, during advanced AD stages, chronic inflammatory microenvironments induce microglial dysfunction with M1 phenotype predominance. Activated M1 microglia release IL-1β and IL-18 through NLRP3 inflammasome-dependent mechanisms. These pro-inflammatory cytokines not only directly damage neurons but also drive astrocyte transformation into neurotoxic A1 phenotypes (35,60). Concurrent C3 complement secretion mediates aberrant synaptic pruning via C3aR-C3a axis activation, significantly reducing hippocampal synaptic density (60,61). Reactive nitrogen species such as nitric oxide (NO) produced by M1 microglia modify Tau proteins, increasing phosphorylation levels and neurofibrillary tangle formation (62,63). Neuropathological analyses reveal positive correlations between cerebrospinal fluid (CSF) M1 markers (e.g., TREM2, IL-6) and Tau-PET signal intensity in patients with AD, suggesting pro-inflammatory microglia accelerate Tau pathology propagation (37,64). In late-stage disease, aging-associated lysosomal acidification defects prevent effective Aβ degradation, leading to intracellular lipofuscin-like inclusion accumulation and autophagic-lysosomal pathway suppression (45,65). Sustained Aβ oligomer exposure induces endoplasmic reticulum stress, activating the unfolded protein response and triggering CHOP-dependent apoptosis, thereby depleting protective microglial populations (66,67). Transcriptomic profiling reveals functional exhaustion characterized by downregulated NFE2 like bZIP transcription factor 2 antioxidant and peroxisome proliferator activated receptor (PPAR)γ pathways (44).
Lipid metabolism as a regulator of microglial function
As illustrated in Fig. 2, microglial functional states critically depend on dynamic lipid metabolic homeostasis. Lipids not only constitute fundamental membrane structures but also act as signaling molecules, energy substrates and inflammatory mediators, deeply engaging in phagocytosis, inflammatory responses and tissue repair processes (48,68). Lipid metabolic dysregulation is closely associated with neurodegenerative pathologies, such as AD (48,68). Microglia sense extracellular lipid microenvironmental changes through specialized receptors, integrating lipid signals via endocytic, oxidative, synthetic and storage pathways to modulate functional phenotypes (69,70).
Microglia detect parenchymal lipid composition changes through multiple PRRs. CD36, a class B scavenger receptor, recognizes oxidized low-density lipoprotein and Aβ while facilitating fatty acid transmembrane transport (70,71). CD36 activation triggers MyD88-dependent signaling via TLR4/6 complexes, promoting pro-inflammatory cytokine release (e.g., IL-1β, TNF-α) and enhancing phagocytic activity (72,73). In AD models, CD36-Aβ interactions drive NLRP3 inflammasome assembly, accelerating amyloid deposition and neuronal damage (30,74). APOE regulates microglial lipid/Aβ uptake through TREM2 receptor binding. The APOE4 isoform exhibits impaired lipid-loading capacity, causing cholesterol clearance defects that elevate AD risk (20,75). Of note, APOE competes with Aβ for TREM2 binding, with APOE dysfunction synergistically promoting Tau pathology spread (76,77).
Microglial lipid handling involves intricate enzymatic cascades and organelle crosstalk. Lipoprotein lipase maintains membrane phospholipid homeostasis and supports phagocytosis by hydrolyzing triacylglycerols to release free fatty acids (FFAs) (78,79). Lysosomal acid lipase (LAL) degrades internalized lipoproteins, with LAL deficiency causing cholesteryl ester (CE) accumulation and lysosomal storage disorders (80). During myelin clearance, microglia recognize phosphatidylserine on apoptotic cells via MER proto-oncogene, tyrosine kinase receptors, initiating phagolysosomal pathways (81,82). Myelin-derived FFAs activate nuclear receptors LXR and PPARγ, inducing cholesterol efflux transporter ATP binding cassette subfamily A member 1 (ABCA1) expression to promote reverse cholesterol transport and suppress inflammation (83,84). However, aging or chronic pathology induces myelin debris overload exceeding microglial clearance capacity, leading to lipid droplet (LD) formation and mitochondrial dysfunction that drive pro-inflammatory polarization (85,86). Preclinical studies have shown that the PPARγ agonist pioglitazone exhibits reproducible neuroprotective effects in tissue culture and animal models (87). However, clinical trials of Pioglitazone in early PD have indicated that it is ineffective, possibly due to toxin-induced animal models not reflecting the pathogenesis of PD or pioglitazone failing to reach the target nigral neurons and not achieving sufficient drug exposure in clinical trials (88). Peroxisomal β-oxidation degrades very-long-chain fatty acids, with its dysfunction causing lipotoxic mediator accumulation (e.g., ceramides) that induce oxidative stress and aberrant inflammatory signaling (89,90). Multifunctional protein-2-deficient mice exhibit microglial metabolic reprogramming characterized by enhanced glycolysis, reactive oxygen species (ROS) bursts and pro-inflammatory cytokine hypersecretion (91,92). Mitochondrial β-oxidation correlates with anti-inflammatory phenotypes, showing significant upregulation in alternatively activated microglia to support tissue repair through ATP generation (93). However, pro-inflammatory stimuli disrupt mitochondrial TCA cycling, causing the accumulation of citrate, which is exported to the cytoplasm for acetyl-CoA conversion, driving de novo lipogenesis (DNL) and LD biogenesis (94,95). This metabolic shift provides precursors for inflammatory mediator synthesis (e.g., prostaglandins) while altering membrane fluidity to affect receptor signaling (96,97).
Global lipidome remodeling drives microglial phenotypic switching. Pro-inflammatory activation enhances glycolysis and mitochondrial fragmentation, inducing citrate efflux that activates ATP-citrate lyase to promote acetyl-CoA production and DNL initiation (94,98). Endoplasmic reticulum-resident fatty acid elongases [e.g., ELOVL fatty acid elongase 5 (ELOVL5)] and desaturases (e.g., stearoyl-CoA desaturase) modify nascent FFAs for membrane phospholipid integration or triacylglycerol (TAG) storage (97,99). LDs serve dual roles as neutral lipid reservoirs sequestering arachidonic acid to limit peroxidation, while functioning as prostaglandin synthesis platforms regulating inflammatory cascades (100,101). In aged brains, lipid droplet-accumulating microglia exhibit TAG enrichment, phagocytic impairment and pro-inflammatory mediator hypersecretion, emerging as neurodegenerative drivers (102). Conversely, anti-inflammatory phenotypes activate LXR/PPARγ signaling to enhance cholesterol efflux and restore remyelination capacity (103,104).
Dysregulated lipid metabolism in neurodegeneration
AD
The human brain, being the most lipid-rich organ, maintains complex lipid composition with exquisite homeostatic regulation. Patients with AD exhibit significant lipidomic alterations across multiple brain regions (105-109). Fatty acid metabolic dysregulation-observed even in early AD-manifests as elevated total FFAs in CSF with subtype-specific variations: ω-3 polyunsaturated fatty acids (PUFAs) like docosahexaenoic acid decrease markedly in vulnerable regions, while pro-inflammatory arachidonic acid levels rise abnormally (110-112). This PUFA/saturated fatty acid ratio imbalance not only reduces membrane fluidity but may also disrupt amyloid precursor protein (APP) processing via lipid raft structural modifications, thereby promoting Aβ generation (113-115). Notably, decreased ω-3 PUFAs/monounsaturated fatty acids and reduced desaturation indices in lipid rafts emerge early in entorhinal and frontal cortices, suggesting lipid microenvironment alterations precede classical pathological deposition (116).
Sphingolipid dysregulation constitutes another core feature of AD lipid abnormalities, with ceramide accumulation strongly correlating with neurodegeneration. Patients with AD show elevated ceramide levels alongside decreased sphingomyelin in prefrontal and temporal cortices, associated with enhanced sphingomyelinase activity and downregulated synthetic enzymes (117-121). Ceramide accumulation exacerbates neuronal damage through mitochondrial oxidative stress and apoptosis while directly promoting Aβ production via β-secretase activation (122-124). Myelin-specific lipids (e.g., sulfatides, galactosylceramides) exhibit early depletion in AD, potentially linked to oligodendrocyte dysfunction and white matter integrity loss-pathological changes confirmed to correlate with cognitive decline (125-127). Despite blood-brain barrier restrictions, AD brains demonstrate elevated cholesterol levels positively correlating with pathological severity (118,128-130). Enhanced acetyl-CoA acetyltransferase 1 (ACAT1) activity in the entorhinal cortex promotes CE accumulation, which exacerbates Aβ deposition through APP amyloidogenic cleavage and alters γ-secretase localization via lipid raft modulation (131-133). Notably, astrocyte-specific cholesterol synthesis inhibition significantly alleviates Aβ and Tau pathology in AD mice, indicating glial cholesterol metabolism exerts transcellular regulatory effects (134).
Genetic studies underscore the central role of lipid metabolism genes in AD risk. APOEε4-the strongest genetic risk factor-impairs lipid transport alongside Aβ clearance: APOEε4 carriers exhibit reduced lipoprotein lipidation compared to APOEε3, leading to defective neuronal lipid clearance and mitochondrial dysfunction (135,136). Risk genes like TREM2 and ABCA1/ABCA7 influence AD progression by regulating microglial lipid phagocytosis/efflux. The TREM2 R47H variant loses CE recognition capacity, causing lipid droplet dysmetabolism and impaired remyelination (15,76,137,138). These findings collectively implicate lipid trafficking defects as multi-mechanism drivers of AD pathology. AD brains show reduced pro-resolving mediators like DHA-derived resolvins alongside elevated pro-inflammatory prostaglandin E2, perpetuating chronic neuroinflammation through pro-/anti-inflammatory mediator imbalance (139-143). Aberrant lipid sensing in glial cells exacerbates this dysregulation: APOE4 astrocytes exhibit impaired fatty acid β-oxidation, causing intracellular lipid accumulation and pro-inflammatory cytokine release (135). Ceramides directly activate NLRP3 inflammasomes, establishing lipid-inflammation positive feedback loops that accelerate neuronal injury (144,145).
PD
The neuropathological hallmark of PD is the formation of Lewy bodies (LBs), proteinaceous inclusions composed of abnormally aggregated α-syn. Studies have revealed that dysregulated lipid metabolism not only modulates α-syn pathology but also interacts with mitochondrial dysfunction, endoplasmic reticulum stress and inter-organelle communication, collectively forming a complex pathogenic network in PD. Notably, lipid metabolic disturbances-particularly the dyshomeostasis of PUFAs, cholesterol and cardiolipin-play pivotal roles in driving dopaminergic neurodegeneration.
Lipid accumulation observed in patients with PD is closely linked to α-syn aggregation (146). Mutations in the glucosylceramidase β1 (GBA1) gene represent one of the most significant genetic risk factors for PD. These mutations impair lysosomal function, leading to intracellular cholesterol and PUFA accumulation (147). Elevated PUFAs not only upregulate α-syn gene expression but also directly induce oligomerization of α-syn monomers into neurotoxic intermediates (148). Lipid-mediated conformational changes in α-syn may involve interactions with cellular membranes: Binding to cholesterol-rich lipid rafts accelerates the transition of soluble α-syn monomers into insoluble fibrils, providing a molecular basis for LB formation (149,150). Furthermore, elevated ApoE levels in the cerebrospinal fluid of patients with early-stage PD suggest that α-syn exploits ApoE-mediated lipid transport systems for interneuronal pathological propagation, amplifying the impact of lipid dysregulation in PD progression (151).
Mitochondrial-ER crosstalk emerges as a critical pathological axis in PD. Excessive fatty acid accumulation triggers mitochondria-cytoplasmic stress responses, generating ROS that preferentially oxidize PUFAs. This lipid peroxidation cascade alters mitochondrial membrane permeability and disrupts electron transport chain function (152). Loss of ER-localized very-long-chain fatty acid elongase Elovl2 disrupts fatty acid synthesis, leading to short-chain fatty acid accumulation within the ER lumen. This metabolic perturbation induces sustained ER stress through unfolded protein overload, culminating in mitochondrial decompensation, exacerbating ROS production and establishing a vicious cycle of mitochondrial decline (153,154). Cardiolipin (CL), a signature phospholipid of mitochondrial inner membranes, exhibits complex pathological interactions in PD. Misfolded α-syn directly binds CL, compromising mitochondrial membrane integrity and impairing oxidative phosphorylation (155). Critically, CL governs mitophagy quality control: Aberrant CL levels impair selective clearance of damaged mitochondria while activating NLRP3 inflammasomes and the cGAS-cGAMP-STING-TBK1-interferon regulatory factor 3 pathway, synergistically exacerbating dopaminergic neuron degeneration (156,157).
Microglia bridge lipid dysregulation and neurodegeneration in PD by sensing metabolic disturbances. Oxidized lipids and glycosphingolipids activate microglia, triggering TNF-α and IL-1β release and establishing chronic neuroinflammation, particularly in GBA1-mutant PD models (158-160). Elevated glycoprotein NMB protein levels in PD substantia nigra microglia correlate with cerebral lipid accumulation, indicating lipid metabolic stress directly drives neuroimmune responses (161). The ApoE ε4 allele exacerbates microglial clearance deficits by impairing lipid transport, facilitating α-syn pathology spread (162,163). Notably, lipopolysaccharide (LPS)-induced microglial activation enhances neuronal susceptibility to α-syn aggregation (164,165). Dysregulated neuron-astrocyte lipid metabolic coupling further contributes to PD: Neurons export toxic fatty acids via ApoE-positive lipid droplets to astrocytes for mitochondrial β-oxidation. However, α-syn-induced astrocytic mitochondrial dysfunction disrupts this detoxification pathway, leading to toxic lipid reaccumulation in neurons and establishing a PD-accelerating feedback loop (166,167).
Other diseases
In HD, synaptic membrane cholesterol dyshomeostasis precedes myelin structural alterations. Mutant huntingtin directly disrupts cholesterol biosynthesis by suppressing sterol regulatory element-binding proteins, leading to significantly reduced total cholesterol levels in neurons and brain tissues of patients with HD and transgenic animal models (168-172). Dynamic changes in 24-hydroxycholesterol (24-OHC) reveal stage-specific metabolic imbalances: Transient elevation in early disease stages reflects compensatory mechanisms, while progressive decline in later stages correlates with neuronal degeneration and attenuated LXR signaling, with plasma 24-OHC levels strongly associated with disease progression (171,173,174). Gene expression analyses demonstrate sustained downregulation of key cholesterol synthesis enzymes (e.g., 3-hydroxy-3-methylglutaryl-CoA reductase, cytochrome P450 family 51) in cortical tissues of patients with HD and in animal models of HD, positively correlating with CAG repeat length and mHTT burden (172,175-177). Astrocytic dysfunction exacerbates metabolic disturbances, characterized by reduced ApoE secretion, while LXR agonists partially rescue these phenotypes, highlighting the central role of glia-neuron crosstalk (171,178,179). Patients with HD also exhibit sphingolipid dysregulation, including elevated total ceramides but marked reductions in myelin-critical dihydroceramide species (e.g., C18:0). Regional deficiencies in ganglioside GM1 and sulfatides further indicate impaired synthesis pathways, destabilizing myelination across brain regions (180).
ALS is intricately linked to systemic lipid metabolic disturbances. Blood profiling reveals significant elevations in plasma sphingomyelins, ceramides and triglycerides in patients with ALS, with a 32-metabolite panel (including 11 lipid species) effectively distinguishing patients from healthy controls (181-185). Blood-brain barrier permeability alterations may exacerbate central-peripheral lipid exchange abnormalities (186,187). Elevated phosphatidylcholine (PC 36:4) in CSF and its recapitulation in superoxide dismutase 1 (SOD1)-G93A mouse brain tissues suggest direct involvement of central lipid remodeling in ALS pathogenesis (188,189).
Postmortem studies demonstrate pronounced lipid abnormalities in ALS spinal cords, including increased triglycerides and lysophosphatidylcholine in gray matter, with ceramide, sphingomyelin and complex sphingolipids accumulating in correlation with disease severity (190-192). Animal models mirror these findings: SOD1 mutant mice exhibit progressive ceramide and CE accumulation in spinal tissues, while fused in sarcoma-overexpressing mice display cardiolipin downregulation and altered nuclear membrane lipid composition (190,192-195). Aberrant ceramide accumulation in ALS correlates with oxidative stress-driven enhancement of de novo sphingolipid synthesis, with dysregulated serine palmitoyltransferase activity due to SPTLC1 variants implicated in familial ALS (190,196,197). CE accumulation in ALS patient spinal cords and SOD1 models positively correlates with disease progression, and its byproduct lysoPC directly damages motor neurons (191). Although serum cholesterol levels show inconsistent associations with ALS prognosis, CNS-specific oxysterols (e.g., 25-OHC) exhibit validated neurotoxicity in cellular and animal models (198,199).
Lipid homeostasis and microglial-mediated neuroinflammation
Lipid droplet accumulation and microglial dysfunction
Cerebral metabolism is tightly regulated to ensure precise neuronal function (200). Although the brain primarily relies on circulating glucose for energy, lipids also support neuronal activity through metabolic crosstalk with astrocytes (166,201). A study has revealed lipid-laden vesicles in glial cells of patients with dementia, with lipid-accumulating microglia exhibiting functional impairments, suggesting a critical link between microglial function and lipid dynamics (202). Abnormal lipid accumulation, particularly lipid dropletscytoplasmic organelles enriched in lipids that regulate cellular lipid metabolism and immune responses-is a key driver of myeloid cell immune dysfunction (203). LDs are bounded by a monolayer membrane decorated with structural proteins, including perilipin family proteins and lipid-metabolizing enzymes, which are essential for maintaining LD morphology and function. Microglial LDs are strongly implicated in neurodegenerative disease pathogenesis (204). In aged mouse models, LD-rich microglia display functional dysregulation, characterized by elevated ROS, increased pro-inflammatory cytokine production and impaired phagocytosis (102). Research in Grn (progranulin-encoding gene) knockout mice demonstrated that 9-month-old mice exhibit pronounced LD accumulation in hippocampal microglia. Transcriptomic profiling revealed that LD-enriched microglia in Grn-/-mice share gene network signatures with 20-month-old wild-type aged mice, indicating conserved lipid-associated dysfunction in neurodegeneration and aging (205). In a cerebral ischemia rat model, Lin et al (206) observed that serum glucose-oxygen deprivation significantly increases microglial LD accumulation, concurrent with elevated release of inflammatory mediators TNF-α and IL-1β. Pharmacological inhibition of LD formation not only reduced microglial inflammatory activation and mortality but also attenuated cerebral infarct volume and motor deficits in ischemic rats (206). Similarly, in a kainic acid-induced hippocampal CA1 injury model, perilipin-2 (an LD surface protein) colocalized with the microglial marker Iba1. These LD-laden microglia exhibited Apoc2 (a neutral lipid-associated apolipoprotein) deposits colocalized with lysosomal marker Lamp1, accompanied by phagocytic impairment and ROS-mediated neuronal damage (207). Collectively, these studies demonstrate that aberrant LD accumulation in microglia represents a hallmark of dysfunctional, pro-inflammatory microglial states in the aging brain and is a consistent feature across neuroimmune-associated neurodegenerative pathologies.
Cholesterol dynamics and microglial functional impairments
Early neuropathological studies identified cytoplasmic neutral lipid inclusions within glial cells in AD patient brains (202). Subsequent investigations have delineated the pathological significance of CE accumulation, observed in AD postmortem specimens (208) and recapitulated across multiple neurodegenerative disease models (208). In vitro evidence demonstrates that ACAT1-mediated cholesterol esterification converts myelin-derived cholesterol into CE for storage within LDs (as shown in Fig. 3). Pharmacological or genetic inhibition of this enzyme exerts neuroprotective effects in Aβ and tau pathology models through autophagy activation (209), indicating ACAT1's regulatory role in disease progression via CE metabolism. Endolysosomal cholesterol accumulation has been observed in APOE4-homozygous human induced pluripotent stem cell (iPSC)-derived astrocytes (210), implicating APOE4-induced glial cholesterol dyshomeostasis in late-onset AD pathogenesis. Three human APOE isoforms (APOE2, APOE3, APOE4), encoded by distinct alleles on chromosome 19 (211), exhibit differential lipid-binding capacities. APOE4 demonstrates reduced cholesterol efflux efficiency, predisposing to intracellular LD accumulation (212). Comparative analyses reveal more pronounced LD deposition in APOE4-expressing microglia vs. APOE3 counterparts (213). iPSC studies further show APOE4 upregulates sterol regulatory element-binding protein 2 to drive de novo cholesterol synthesis while disrupting cholesterol trafficking via lysosomal sequestration of free cholesterol (214). Emerging data indicate APOE4 downregulates mitochondrial oxidative phosphorylation genes and activates lipogenic pathways, exacerbating microglial LD accumulation (215). Reduced cholesterol uptake by APOE4 microglia increases extracellular cholesterol incorporation into neuronal membranes, enhancing lipid-gated potassium channel activity (212). Pharmacological inhibition of acyl-CoA synthetase 1 (ACSL1) to modulate LD levels restores purinergic signaling and sustains microglial homeostasis, supporting neural network integrity (215).
Oxysterols, cholesterol-derived metabolites, impair microglial clearance of cellular debris and neurotoxic molecules through direct cytotoxicity or downstream pathway modulation. Deficient Aβ monomer clearance promotes oligomerization and plaque formation-cardinal AD pathological hallmarks (216,217). Enzymatic conversion of cholesterol by 25-hydroxylase and 27-hydroxylase generates 25-OHC and 27-OHC, respectively, while 7-ketocholesterol arises via ROS-mediated non-enzymatic oxidation (218). Experimental data demonstrate that 25-OHC synergizes with LPS to induce inducible NO synthase (iNOS) mRNA expression (219), and 27-OHC activates the IL-6/STAT3 pro-inflammatory axis (220). ROS-driven accumulation of non-enzymatic oxysterols sustains chronic disease-associated microglial activation (221). Mechanistically, 25-OHC increases membrane permeability through lipid bilayer disorganization, triggering cell death (222-224). Concurrently, 27-OHC induces senescence via oxidative damage (225), while 7-ketocholesterol disrupts peroxisomal activity and mediates activated microglia-induced neuronal injury through oxidative stress (226).
LD accumulation driven by cholesterol dysmetabolism correlates with pro-inflammatory lipidomic signatures (227). LXR activation suppresses CNS inflammation, enhances lipid recycling and improves microglial function in rodent models (228). Cholesterol-enriched membranes promote lipid raft assembly, facilitating TLR dimerization and amplifying LPS-induced pro-inflammatory signaling (229). Chronic LPS exposure exacerbates Aβ deposition (230), while a 3% high-cholesterol diet induces inflammasome activation and blood-brain barrier disruption via pro-inflammatory microglial polarization (231). APOE4 carriers exhibit heightened neuroinflammation under hypercholesterolemic conditions due to impaired cholesterol efflux (232), highlighting genetic susceptibility to chronic microglial activation. Elevated CNS cholesterol levels inhibit phagocytic capacity (75), whereas methyl-β-cyclodextrin-mediated cholesterol depletion enhances phagocytosis (233). Of note, CE accumulation caused by TREM2 deficiency does not impair phagocytic function (20), underscoring the primacy of cholesterol transport efficiency over total cholesterol load. ABCA1-deficient microglia exhibit increased TNF-α secretion and reduced phagocytosis (234), while ABCA7 loss accelerates amyloid precursor protein processing and impairs Aβ clearance (235). Translocator protein knockout AD models display exacerbated Aβ deposition alongside diminished phagocytic microglia (236), validating functional phagocytosis as a critical preventive mechanism in AD.
Microglial regulation of demyelination and remyelination
Demyelination contributes to neuronal dysfunction and cognitive decline in AD (237). Clinical studies demonstrate oligodendrocyte degeneration and reduced/damaged phospholipid content in AD brains (238,239). Single-cell transcriptomic analyses reveal significant gene expression alterations in oligodendrocyte lineage cells during early AD stages (240), with myelination-related pathways consistently disrupted across multiple cell types in prefrontal cortical tissues, underscoring myelination's pivotal role in AD pathophysiology (240). Microglia critically maintain myelin integrity. Under homeostatic conditions, axonal myelination is dynamically regulated by a demyelination-remyelination equilibrium requiring coordinated interactions between oligodendrocyte progenitor cells (OPCs) and microglia (241). Microglia clear myelin debris and secrete signaling factors to promote OPC differentiation into oligodendrocytes (242). However, aging and neuropathological accumulation accelerate remyelination decline, impairing myelin maintenance and causing myelin debris accumulation (85). Excessive debris disrupts OPC recruitment to axons and suppresses microglia-derived factors essential for OPC differentiation, further compromising remyelination (241). While microglia protectively degrade phagocytosed myelin debris via autophagy and secrete pro-remyelination cytokines (9,243), their clearance capacity is finite. Pathological myelin overload exceeding lysosomal degradation capacity induces lysosomal damage, triggering microglial senescence and pro-inflammatory cytokine release-termed the senescence-associated secretory phenotype (SASP) (85). SASP microglia exacerbate pathology through dual mechanisms: Impaired phagocytosis amplifies amyloid plaque and myelin debris accumulation, while debris-activated microglia directly drive neuroinflammation, damaging neurons, oligodendrocytes and OPCs (241).
Similar to astrocytes, microglia/macrophages exhibit context-dependent dual activation states: Pro-inflammatory classical (M1; marked by iNOS, TNF, CD16/CD32) and anti-inflammatory pro-regenerative (M2; expressing Arg1, insulin-like growth factor 1, CD206). Focal lysolecithin-induced corpus callosum demyelination models demonstrate M1-to-M2 phenotypic switching during remyelination initiation, involving necroptosis of pro-inflammatory subsets and repopulation by regenerative phenotypes (244,245). In vitro, M2-conditioned media enhances OPC differentiation, abolished by M2 depletion in lesions. Heterochronic parabiosis (a surgical model that connects the circulatory systems of young and old mice) shows aged mice exhibit increased lesion M2 cells with enhanced remyelination, blocked by inhibiting M2-derived activin-A (244). Microglial activation transcends simplistic M1/M2 dichotomies, encompassing broader phenotypic spectra. In Theiler's virus-induced demyelination, endocannabinoid 2-arachidonoylglycerol induces microglial phagocytosis and OPC differentiation without altering classical M1/M2 markers (246). Myelin debris induces novel foamy macrophage phenotypes with ABCA1 deficiency, impairing apoptotic/necrotic cell clearance and exacerbating secondary injury (247). Single-cell RNA sequencing in experimental autoimmune encephalomyelitis (EAE) mice reveals shared activation markers (CD44, CD86, programmed cell death ligand 1) alongside Major histocompatibility complex class II/stem cell antigen-1 upregulation and CD14 downregulation (248). Lysolecithin lesion microglia form distinct subsets expressing APOE but diverging in CCL4/CXCL10 expression (249). Demyelination models identify APOE-dependent molecular signatures overlapping with those of microglia from patients with MS (48,250). Real-time imaging captures dynamic microglial/macrophage shifts from pro-inflammatory to immunoregulatory states in EAE lesions (251). CD11c+ microglia-critical for postnatal myelination-expand in EAE and cuprizone models. Colony stimulating factor 1 receptor activation drives CD11c+ expansion via C-C motif chemokine ligand 2 chemotaxis, ameliorating EAE and reducing demyelination (252). The homeobox gene muscle segment homeobox 3 regulates polarization by oppositely modulating PPARγ/STAT6/JAK3 in M2/M1 cells, influencing disease progression across models (253). During EAE recovery, pro-regenerative microglia upregulate purinergic receptor P2X4R, whose activation enhances debris clearance and suppresses inflammation-effects abolished by receptor blockade (254).
Ferroptosis linking lipid metabolism and neuroinflammation
The interplay between dysregulated lipid metabolism in microglia and ferroptosis constitutes a critical pathological mechanism underlying neurodegenerative disorders (255). Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, particularly depends on the aberrant oxidation of phospholipids enriched with PUFAs (256). As resident immune sentinels in the CNS, microglial susceptibility to ferroptosis and their involvement in neuroinflammatory processes are fundamentally regulated by their polarization states through lipid metabolic reprogramming (102). At the molecular level, the 15-lipoxygenase (15-LOX)/phosphatidylethanolamine-binding protein 1 complex serves as a pivotal driver of ferroptosis (257). This enzymatic complex specifically catalyzes the peroxidation of arachidonoyl-phosphatidylethanolamine to generate 15-hydroperoxy-eicosatetraenoyl-phosphatidylethanolamine (15-HpETE-PE) (as shown in Fig. 4), a potent ferroptosis-inducing lipid species (258). This process requires both redox-active iron-generated ROS and precise enzymatic regulation. Of note, structural studies revealed that 15-LOX contains specific 'oxygen channels' permitting competitive access of molecular oxygen and NO radicals (NO•), thereby dynamically modulating lipid peroxidation kinetics (259). Polarization status critically determines microglial ferroptosis susceptibility. Pro-inflammatory M1 microglia exhibit ferroptosis resistance through iNOS-mediated NO• production, which directly binds to the Fe2+ catalytic center of 15-LOX to inhibit 15-HpETE-PE synthesis (260). Conversely, anti-inflammatory M2 microglia lacking iNOS expression demonstrate impaired 15-LOX inhibition, leading to progressive lipid peroxide accumulation and eventual ferroptotic death (260). Beyond the LOX system, Acyl-coA synthetase long chain family member 4 (ACSL4) exacerbates ferroptotic vulnerability by esterifying PUFAs into membrane phospholipids, increasing oxidizable substrate availability (261). Experimental evidence from sepsis-associated encephalopathy models demonstrates that ACSL4 upregulation mediates neuroinflammation through NF-κB pathway activation, while pharmacological inhibition with propofol attenuates hippocampal ferroptosis by ACSL4 downregulation, preserving neuronal viability, as evidenced by Nissl staining and reduced Fluoro-Jade C positivity (262,263). These findings position ACSL4 as a key regulator of microglial membrane lipid composition and neurodegeneration progression. The glutathione peroxidase 4 (GPX4) antioxidant system provides crucial protection against ferroptosis by reducing lipid peroxides to non-toxic alcohols using reduced glutathione (264). Importantly, experimental models of sepsis-associated encephalopathy have revealed significant GPX4 downregulation in affected brain regions (265). Notably, aging-associated microglial dysfunction involves heme oxygenase-1 upregulation, which suppresses GPX4 expression and promotes iron accumulation, driving M1 polarization and neurotoxic cytokine production (IL-1β, TNF-α, NLRP3, caspase-1) (266). Therapeutic interventions with ferroptosis inhibitors demonstrate dual benefits by scavenging free radicals, suppressing lipid peroxidation and promoting microglial phenotype switching from pro-inflammatory M1 to neuroprotective M2 states, accompanied by enhanced phagocytic capacity and reduced inflammatory mediator secretion (267). Collectively, these mechanisms establish microglial lipid metabolic dysregulation as a central hub integrating ferroptotic cell death with neuroinflammatory cascades in neurodegenerative pathogenesis.
Therapeutic advances targeting lipid metabolism
TREM2 as a lipid metabolism regulatory target in NDDs
Modulating lipid metabolism pathways shows therapeutic potential for AD, with dietary interventions attracting particular attention. Preclinical studies confirm that ω-3 PUFA supplements DHA and eicosapentaenoic acid ameliorate AD pathology in postmenopausal women and older individuals with mild cognitive impairment (268,269). Statins, which inhibit cholesterol synthesis, improve cognitive deficits in patients with AD or amnestic mild cognitive impairment and hypercholesterolemia by targeting TLR4 signaling (270,271). The upregulation of TLR4 in TREM2-deficient models (272) suggests statins may specifically regulate neuroinflammation in patients with AD carrying TREM2 mutations. Mechanistically, TREM2-deficient myeloid cells exhibit pathological CE accumulation, reversible via inhibition of ACAT1 (20), providing a potential therapeutic target for TREM2 mutation carriers (209). Given the loss-of-function nature of most pathogenic TREM2 variants, agonistic antibodies AL002a or engineered TREM2-expressing myeloid cell transplantation are emerging therapeutic approaches (15). Price et al (273) demonstrated that AL002a antibody treatment significantly reduces Aβ pathology and improves cognition in 5XFAD mice. TREM2 also modulates APOE isoform-specific effects: TREM2 deficiency exacerbates microglial Aβ clearance deficits in APOE ε4 mice (274), supporting TREM2-targeted therapies for APOE ε4-associated AD. The RXR agonist bexarotene exhibits multi-target efficacy by enhancing microglial phagocytosis and regulating the TREM2-TYRO protein tyrosine kinase binding protein-CD33 network (275). Transcriptomic analyses reveal bexarotene's dual actions in APP/PS1 mice: Activating immune pathways for Aβ clearance while suppressing neuroinflammation (276). Notably, bexarotene corrects lipoprotein metabolism and cognitive deficits in humanized APOE4 mice (277), suggesting TREM2-mediated APOE isoform regulation underlies its therapeutic effects, though mechanistic details require further elucidation.
Therapeutic potential of fatty acid synthase inhibitors
CMS121, a flavonoid derivative of fisetin, combines neuroprotective and metabolic regulatory properties (278). This compound concurrently inhibits two programmed cell death pathways: Glutathione depletion-associated oxytosis and iron-dependent ferroptosis (279,280). In vitro studies demonstrate CMS121's selective protection against diverse cytotoxic agents: It blocks RSL3-induced lipid peroxidation in HT22 neurons and mitigates glutamate-and erastin-induced cell death in AD mice (280). CMS121 broadly modulates LPS-stimulated microglial responses in BV2 cells, suppressing lipid peroxidation and downregulating pro-inflammatory mediators, including iNOS, TNF-α and cyclooxygenase 2 (280). These effects phenocopy fatty acid synthase (FASN) gene silencing, confirming target specificity. In AD mouse models, CMS121 significantly reduces hippocampal 4-hydroxynonenal, a lipid peroxidation marker linked to neuronal damage (281), while decreasing glial fibrillary acidic protein expression, indicating attenuated glial activation (282). CMS121 exerts multi-directional lipid metabolic regulation by lowering endocannabinoids, free fatty acids and PUFAs in AD mice (280). However, it paradoxically elevates ceramide levels-a consequence of FASN inhibition-induced malonyl-CoA accumulation, which blocks carnitine palmitoyltransferase 1-mediated fatty acid β-oxidation and diverts metabolic flux toward ceramide synthesis in brains from patients who suffered from neuropathological disorders (117).
Conclusions and future perspectives
NDDs, including AD, PD and ALS, present significant global health challenges due to progressive neuronal degeneration and associated cognitive and functional impairments. Despite differences in clinical manifestations, these disorders share common pathological mechanisms characterized by neuroinflammation, microglial dysfunction and disrupted lipid metabolism. Microglia, as resident immune cells in the CNS, play critical roles in neurodegenerative disease progression through their dynamic polarization into pro-inflammatory and anti-inflammatory phenotypes (8,9). The M1 phenotype exacerbates neuronal injury through the secretion of inflammatory cytokines, lipid peroxidation and promotion of ferroptosis (44), whereas the M2 phenotype mediates neuroprotective effects by enhancing pathological protein clearance, secreting neurotrophic factors and supporting synaptic plasticity (46,47). Lipid metabolism has emerged as a pivotal factor influencing microglial function and neuroinflammatory processes. Dysregulated cholesterol metabolism and fatty acid oxidation, mediated by key molecules such as TREM2 and ACAT1, lead to lipid droplet accumulation and pro-inflammatory activation in microglia (138,209). Furthermore, lipid peroxidation products and ferroptotic pathways, driven by altered lipid metabolism, establish crucial mechanistic links between microglial dysfunction and neuronal cell death (261). Targeting lipid metabolic pathways represents an innovative therapeutic approach for NDDs. Pharmacological agents including TREM2 agonists, ACAT1 inhibitors and fatty acid synthase inhibitors like CMS121 have demonstrated promising efficacy in preclinical studies by reducing neuroinflammation, restoring lipid homeostasis and improving cognitive outcomes (15,281). Future research directions should focus on clarifying the molecular interactions between lipid metabolism and microglial polarization, identifying reliable lipid biomarkers for disease progression and developing optimized lipid-targeted therapeutic interventions. Advances in this field hold significant potential for improving treatment efficacy and establishing novel therapeutic strategies for NDDs.
Availability of data and materials
Not applicable.
Authors' contributions
YS and LG provided the theoretical foundation and wrote the manuscript. YS generated the figures. XL performed the literature search. KW and JW categorized and organized the literature. BP reviewed the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Acknowledgments
Not applicable.
Funding
This review was funded by a project supported by the Natural Science Foundation of Shandong Province, China (grant no. ZR2024QH109) and the Youth Innovation Technology Project of Higher School in Shandong Province, China (grant no. 2023KJ264).
References
Lamptey RNL, Chaulagain B, Trivedi R, Gothwal A, Layek B and Singh J: A Review of the common neurodegenerative disorders: Current therapeutic approaches and the potential role of nanotherapeutics. Int J Mol Sci. 23:18512022. View Article : Google Scholar : PubMed/NCBI | |
Hou Y, Dan X, Babbar M, Wei Y, Hasselbalch SG, Croteau DL and Bohr VA: Ageing as a risk factor for neurodegenerative disease. Nat Rev Neurol. 15:565–581. 2019. View Article : Google Scholar | |
Scheltens P, De Strooper B, Kivipelto M, Holstege H, Chetelat G, Teunissen CE, Cummings J and van der Flier WM: Alzheimer's disease. Lancet. 397:1577–1590. 2021. View Article : Google Scholar | |
Kalia LV and Lang AE: Parkinson's disease. Lancet. 386:896–912. 2015. View Article : Google Scholar : PubMed/NCBI | |
Feldman EL, Goutman SA, Petri S, Mazzini L, Savelieff MG, Shaw PJ and Sobue G: Amyotrophic lateral sclerosis. Lancet. 400:1363–1380. 2022. View Article : Google Scholar : PubMed/NCBI | |
Vaquer-Alicea J and Diamond MI: Propagation of protein aggregation in neurodegenerative diseases. Annu Rev Biochem. 88:785–810. 2019. View Article : Google Scholar | |
Dugger BN and Dickson DW: Pathology of neurodegenerative diseases. Cold Spring Harb Perspect Biol. 9:a0280352017. View Article : Google Scholar : PubMed/NCBI | |
Gao C, Jiang J, Tan Y and Chen S: Microglia in neurodegenerative diseases: Mechanism and potential therapeutic targets. Signal Transduct Target Ther. 8:3592023. View Article : Google Scholar : | |
Kent SA and Miron VE: Microglia regulation of central nervous system myelin health and regeneration. Nat Rev Immunol. 24:49–63. 2024. View Article : Google Scholar | |
Voet S, Srinivasan S, Lamkanfi M and van Loo G: Inflammasomes in neuroinflammatory and neurodegenerative diseases. EMBO Mol Med. 11:e102482019. View Article : Google Scholar : PubMed/NCBI | |
Sun N, Victor MB, Park YP, Xiong X, Scannail AN, Leary N, Prosper S, Viswanathan S, Luna X, Boix CA, et al: Human microglial state dynamics in Alzheimer's disease progression. Cell. 186:4386–4403.e29. 2023. View Article : Google Scholar : PubMed/NCBI | |
Yu H, Chang Q, Sun T, He X, Wen L, An J, Feng J and Zhao Y: Metabolic reprogramming and polarization of microglia in Parkinson's disease: Role of inflammasome and iron. Ageing Res Rev. 90:1020322023. View Article : Google Scholar : PubMed/NCBI | |
Clarke BE and Patani R: The microglial component of amyotrophic lateral sclerosis. Brain. 143:3526–3539. 2020. View Article : Google Scholar | |
Billingham LK, Stoolman JS, Vasan K, Rodriguez AE, Poor TA, Szibor M, Jacobs HT, Reczek CR, Rashidi A, Zhang P, et al: Mitochondrial electron transport chain is necessary for NLRP3 inflammasome activation. Nat Immunol. 23:692–704. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Cella M, Mallinson K, Ulrich JD, Young KL, Robinette ML, Gilfillan S, Krishnan GM, Sudhakar S, Zinselmeyer BH, et al: TREM2 lipid sensing sustains the microglial response in an Alzheimer's disease model. Cell. 160:1061–1071. 2015. View Article : Google Scholar : | |
Zheng Z, Chen M, Feng S, Zhao H, Qu T, Zhao X, Ruan Q, Li L and Guo J: VDR and deubiquitination control neuronal oxidative stress and microglial inflammation in Parkinson's disease. Cell Death Discov. 10:1502024. View Article : Google Scholar | |
Yoon H, Shaw JL, Haigis MC and Greka A: Lipid metabolism in sickness and in health: Emerging regulators of lipotoxicity. Mol Cell. 81:3708–3730. 2021. View Article : Google Scholar : PubMed/NCBI | |
Shang C, Su Y, Ma J, Li Z, Wang P, Ma H, Song J and Zhang Z: Huanshaodan regulates microglial glucose metabolism reprogramming to alleviate neuroinflammation in AD mice through mTOR/HIF-1α signaling pathway. Front Pharmacol. 15:14345682024. View Article : Google Scholar | |
Baik SH, Kang S, Lee W, Choi H, Chung S, Kim JI and Mook-Jung I: A breakdown in metabolic reprogramming causes microglia dysfunction in Alzheimer's disease. Cell Metab. 30:493–507.e6. 2019. View Article : Google Scholar : PubMed/NCBI | |
Nugent AA, Lin K, van Lengerich B, Lianoglou S, Przybyla L, Davis SS, Llapashtica C, Wang J, Kim DJ, Xia D, et al: TREM2 regulates microglial cholesterol metabolism upon chronic phagocytic challenge. Neuron. 105:837–854.e9. 2020. View Article : Google Scholar : PubMed/NCBI | |
Chausse B, Kakimoto PA and Kann O: Microglia and lipids: How metabolism controls brain innate immunity. Semin Cell Dev Biol. 112:137–144. 2021. View Article : Google Scholar | |
Lull ME and Block ML: Microglial activation and chronic neurodegeneration. Neurotherapeutics. 7:354–365. 2010. View Article : Google Scholar | |
Fernando KKM and Wijayasinghe YS: Sirtuins as potential therapeutic targets for mitigating neuroinflammation associated with Alzheimer's disease. Front Cell Neurosci. 15:7466312021. View Article : Google Scholar | |
Licata L, Viale G, Giuliano M, Cur igliano G, Chavez-MacGregor M, Foldi J, Oke O, Collins J, Del Mastro L, Puglisi F, et al: Oncotype DX results increase concordance in adjuvant chemotherapy recommendations for early-stage breast cancer. NPJ Breast Cancer. 9:512023. View Article : Google Scholar : | |
Weiss F, Hughes L, Fu Y, Bardy C, Halliday GM and Dzamko N: Astrocytes contribute to toll-like receptor 2-mediated neurodegeneration and alpha-synuclein pathology in a human midbrain Parkinson's model. Transl Neurodegener. 13:622024. View Article : Google Scholar : PubMed/NCBI | |
Sun L, Jiang WW, Wang Y, Yuan YS, Rong Z, Wu J, Fan Y, Lu M and Zhang KZ: Phosphorylated α-synuclein aggregated in Schwann cells exacerbates peripheral neuroinflammation and nerve dysfunction in Parkinson's disease through TLR2/NF-κB pathway. Cell Death Discov. 7:2892021. View Article : Google Scholar | |
Wendimu MY and Hooks SB: Microglia phenotypes in aging and neurodegenerative diseases. Cells. 11:20192022. View Article : Google Scholar | |
Kettenmann H, Kirchhoff F and Verkhratsky A: Microglia: New roles for the synaptic stripper. Neuron. 77:10–18. 2013. View Article : Google Scholar | |
Platanitis E and Decker T: Regulatory networks involving STATs, IRFs, and NFκB in inflammation. Front Immunol. 9:25422018. View Article : Google Scholar | |
Stewart CR, Stuart LM, Wilkinson K, van Gils JM, Deng J, Halle A, Rayner KJ, Boyer L, Zhong R, Frazier WA, et al: CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer. Nat Immunol. 11:155–161. 2010. View Article : Google Scholar : | |
Wang Y, Wang YC and Ma J: Effects of electroacupuncture on Sirt3/NLRP3/GSDMD signaling pathway in the substantia nigra of midbrain of rats with Parkinson's disease. Zhen Ci Yan Jiu. 49:384–390. 2024.In Chinese. | |
Yu CH, Davidson S, Harapas CR, Hilton JB, Mlodzianoski MJ, Laohamonthonkul P, Louis C, Low RRJ, Moecking J, De Nardo D, et al: TDP-43 triggers mitochondrial DNA release via mPTP to activate cGAS/STING in ALS. Cell. 183:636–649.e18. 2020. View Article : Google Scholar : PubMed/NCBI | |
Becker LA, Huang B, Bieri G, Ma R, Knowles DA, Jafar-Nejad P, Messing J, Kim HJ, Soriano A, Auburger G, et al: Therapeutic reduction of ataxin-2 extends lifespan and reduces pathology in TDP-43 mice. Nature. 544:367–371. 2017. View Article : Google Scholar : PubMed/NCBI | |
Deane R, Singh I, Sagare AP, Bell RD, Ross NT, LaRue B, Love R, Perry S, Paquette N, Deane RJ, et al: A multimodal RAGE-specific inhibitor reduces amyloid β-mediated brain disorder in a mouse model of Alzheimer disease. J Clin Invest. 122:1377–1392. 2012. View Article : Google Scholar : | |
Hanslik KL and Ulland TK: The role of microglia and the Nlrp3 inflammasome in Alzheimer's disease. Front Neurol. 11:5707112020. View Article : Google Scholar : | |
Bolos M, Llorens-Martin M, Perea JR, Jurado-Arjona J, Rabano A, Hernandez F and Avila J: Absence of CX3CR1 impairs the internalization of Tau by microglia. Mol Neurodegener. 12:592017. View Article : Google Scholar : | |
Chidambaram H, Das R and Chinnathambi S: Interaction of Tau with the chemokine receptor, CX3CR1 and its effect on microglial activation, migration and proliferation. Cell Biosci. 10:1092020. View Article : Google Scholar | |
Wilton DK, Mastro K, Heller MD, Gergits FW, Willing CR, Fahey JB, Frouin A, Daggett A, Gu X, Kim YA, et al: Microglia and complement mediate early corticostriatal synapse loss and cognitive dysfunction in Huntington's disease. Nat Med. 29:2866–2884. 2023. View Article : Google Scholar | |
Hong S, Beja-Glasser VF, Nfonoyim BM, Frouin A, Li S, Ramakrishnan S, Merry KM, Shi Q, Rosenthal A, Barres BA, et al: Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science. 352:712–716. 2016. View Article : Google Scholar : | |
Kim A, Garcia-Garcia E, Straccia M, Comella-Bolla A, Miguez A, Masana M, Alberch J, Canals JM and Rodriguez MJ: Reduced fractalkine levels lead to striatal synaptic plasticity deficits in Huntington's disease. Front Cell Neurosci. 14:1632020. View Article : Google Scholar | |
Logan T, Simon MJ, Rana A, Cherf GM, Srivastava A, Davis SS, Low RLY, Chiu CL, Fang M, Huang F, et al: Rescue of a lysosomal storage disorder caused by Grn loss of function with a brain penetrant progranulin biologic. Cell. 184:4651–4668.e25. 2021. View Article : Google Scholar | |
Mata-Martinez E, Diaz-Munoz M and Vazquez-Cuevas FG: Glial cells and brain diseases: Inflammasomes as relevant pathological entities. Front Cell Neurosci. 16:9295292022. View Article : Google Scholar : | |
Xu P, Zhang X, Liu Q, Xie Y, Shi X, Chen J, Li Y, Guo H, Sun R, Hong Y, et al: Microglial TREM-1 receptor mediates neuroinflammatory injury via interaction with SYK in experimental ischemic stroke. Cell Death Dis. 10:5552019. View Article : Google Scholar : | |
Qin Q, Teng Z, Liu C, Li Q, Yin Y and Tang Y: TREM2, microglia, and Alzheimer's disease. Mech Ageing Dev. 195:1114382021. View Article : Google Scholar | |
Caldeira C, Cunha C, Vaz AR, Falcao AS, Barateiro A, Seixas E, Fernandes A and Brites D: Key Aging-associated alterations in primary microglia response to Beta-amyloid stimulation. Front Aging Neurosci. 9:2772017. View Article : Google Scholar : PubMed/NCBI | |
Condello C, Yuan P, Schain A and Grutzendler J: Microglia Constitute a barrier that prevents neurotoxic protofibrillar Aβ42 hotspots around plaques. Nat Commun. 6:61762015. View Article : Google Scholar | |
Feng W, Zhang Y, Wang Z, Xu H, Wu T, Marshall C, Gao J and Xiao M: Microglia Prevent beta-amyloid plaque formation in the early stage of an Alzheimer's disease mouse model with suppression of glymphatic clearance. Alzheimers Res Ther. 12:1252020. View Article : Google Scholar | |
Krasemann S, Madore C, Cialic R, Baufeld C, Calcagno N, El Fatimy R, Beckers L, O'Loughlin E, Xu Y, Fanek Z, et al: The TREM2-APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. Immunity. 47:566–581.e9. 2017. View Article : Google Scholar : | |
Felsky D, Patrick E, Schneider JA, Mostafavi S, Gaiteri C, Patsopoulos N, Bennett DA and De Jager PL: Polygenic analysis of inflammatory disease variants and effects on microglia in the aging brain. Mol Neurodegener. 13:382018. View Article : Google Scholar | |
Absinta M, Maric D, Gharagozloo M, Garton T, Smith MD, Jin J, Fitzgerald KC, Song A, Liu P, Lin JP, et al: A lymphocyte-microglia-astrocyte axis in chronic active multiple sclerosis. Nature. 597:709–714. 2021. View Article : Google Scholar : PubMed/NCBI | |
Werneburg S, Jung J, Kunjamma RB, Ha SK, Luciano NJ, Willis CM, Gao G, Biscola NP, Havton LA, Crocker SJ, et al: Targeted complement inhibition at synapses prevents microglial synaptic engulfment and synapse loss in demyelinating disease. Immunity. 52:167–182.e7. 2020. View Article : Google Scholar : | |
Ellrichmann G, Reick C, Saft C and Linker RA: The role of the immune system in Huntington's disease. Clin Dev Immunol. 2013:5412592013. View Article : Google Scholar : PubMed/NCBI | |
Creus-Muncunill J and Ehrlich ME: Cell-Autonomous and Non-cell-autonomous pathogenic mechanisms in Huntington's disease: Insights from in vitro and in vivo models. Neurotherapeutics. 16:957–978. 2019. View Article : Google Scholar | |
Udeochu JC, Amin S, Huang Y, Fan L, Torres ERS, Carling GK, Liu B, McGurran H, Coronas-Samano G, Kauwe G, et al: Tau activation of microglial cGAS-IFN reduces MEF2C-mediated cognitive resilience. Nat Neurosci. 26:737–750. 2023. View Article : Google Scholar : PubMed/NCBI | |
Hu Y, Fryatt GL, Ghorbani M, Obst J, Menassa DA, Martin-Estebane M, Muntslag TAO, Olmos-Alonso A, Guerrero-Carrasco M, Thomas D, et al: Replicative senescence dictates the emergence of disease-associated microglia and contributes to Aβ pathology. Cell Rep. 35:1092282021. View Article : Google Scholar | |
Yamamoto M, Kiyota T, Walsh SM, Liu J, Kipnis J and Ikezu T: Cytokine-mediated inhibition of fibrillar amyloid-beta peptide degradation by human mononuclear phagocytes. J Immunol. 181:3877–3886. 2008. View Article : Google Scholar | |
Han F, Perrin RJ, Wang Q, Wang Y, Perlmutter JS, Morris JC, Benzinger TLS and Xu J: Neuroinflammation and Myelin Status in Alzheimer's disease, Parkinson's disease, and normal aging brains: A small sample study. Parkinsons Dis. 2019:79754072019.PubMed/NCBI | |
Jurga AM, Paleczna M and Kuter KZ: Overview of general and discriminating markers of differential microglia phenotypes. Front Cell Neurosci. 14:1982020. View Article : Google Scholar : | |
Gadani SP, Cronk JC, Norris GT and Kipnis J: IL-4 in the brain: A cytokine to remember. J Immunol. 189:4213–4219. 2012. View Article : Google Scholar | |
Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, Bennett ML, Munch AE, Chung WS, Peterson TC, et al: Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 541:481–487. 2017. View Article : Google Scholar | |
Tecchio C, Micheletti A and Cassatella MA: Neutrophil-derived cytokines: Facts beyond expression. Front Immunol. 5:5082014. View Article : Google Scholar : PubMed/NCBI | |
Kummer MP, Hermes M, Delekarte A, Hammerschmidt T, Kumar S, Terwel D, Walter J, Pape HC, König S, Roeber S, et al: Nitration of tyrosine 10 critically enhances amyloid β aggregation and plaque formation. Neuron. 71:833–844. 2011. View Article : Google Scholar : PubMed/NCBI | |
Wang WY, Tan MS, Yu JT and Tan L: Role of pro-inflammatory cytokines released from microglia in Alzheimer's disease. Ann Transl Med. 3:1362015.PubMed/NCBI | |
Sheffield LG, Marquis JG and Berman NE: Regional distribution of cortical microglia parallels that of neurofibrillary tangles in Alzheimer's disease. Neurosci Lett. 285:165–168. 2000. View Article : Google Scholar | |
Venegas C and Heneka MT: Danger-associated molecular patterns in Alzheimer's disease. J Leukoc Biol. 101:87–98. 2017. View Article : Google Scholar | |
Jha MK, Jo M, Kim JH and Suk K: Microglia-astrocyte crosstalk: An intimate molecular conversation. Neuroscientist. 25:227–240. 2019. View Article : Google Scholar | |
Hanisch UK and Kettenmann H: Microglia: Active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci. 10:1387–1394. 2007. View Article : Google Scholar | |
Valdearcos M, Douglass JD, Robblee MM, Dorfman MD, Stifler DR, Bennett ML, Gerritse I, Fasnacht R, Barres BA, Thaler JP and Koliwad SK: Microglial inflammatory signaling orchestrates the hypothalamic immune response to dietary excess and mediates obesity susceptibility. Cell Metab. 26:185–197.e3. 2017. View Article : Google Scholar : PubMed/NCBI | |
Doens D and Fernandez PL: Microglia receptors and their implications in the response to amyloid β for Alzheimer's disease pathogenesis. J Neuroinflammation. 11:482014. View Article : Google Scholar | |
Kim E, Tolhurst AT, Qin LY, Chen XY, Febbraio M and Cho S: CD36/fatty acid translocase, an inflammatory mediator, is involved in hyperlipidemia-induced exacerbation in ischemic brain injury. J Neurosci. 28:4661–4670. 2008. View Article : Google Scholar : PubMed/NCBI | |
Gao D, Ashraf MZ, Kar NS, Lin D, Sayre LM and Podrez EA: Structural basis for the recognition of oxidized phospholipids in oxidized low density lipoproteins by class B scavenger receptors CD36 and SR-BI. J Biol Chem. 285:4447–4454. 2010. View Article : Google Scholar : | |
Coraci IS, Husemann J, Berman JW, Hulette C, Dufour JH, Campanella GK, Luster AD, Silverstein SC and El-Khoury JB: CD36, a class B scavenger receptor, is expressed on microglia in Alzheimer's disease brains and can mediate production of reactive oxygen species in response to beta-amyloid fibrils. Am J Pathol. 160:101–112. 2002. View Article : Google Scholar | |
Yamanaka M, Ishikawa T, Griep A, Axt D, Kummer MP and Heneka MT: PPARγ/RXRα-induced and CD36-mediated microglial amyloid-β phagocytosis results in cognitive improvement in amyloid precursor protein/presenilin 1 mice. J Neurosci. 32:17321–17331. 2012. View Article : Google Scholar : PubMed/NCBI | |
Sheedy FJ, Grebe A, Rayner KJ, Kalantari P, Ramkhelawon B, Carpenter SB, Becker CE, Ediriweera HN, Mullick AE, Golenbock DT, et al: CD36 coordinates NLRP3 inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands in sterile inflammation. Nat Immunol. 14:812–820. 2013. View Article : Google Scholar : | |
Cantuti-Castelvetri L, Fitzner D, Bosch-Queralt M, Weil MT, Su M, Sen P, Ruhwedel T, Mitkovski M, Trendelenburg G, Lütjohann D, et al: Defective cholesterol clearance limits remyelination in the aged central nervous system. Science. 359:684–688. 2018. View Article : Google Scholar | |
Yeh FL, Wang Y, Tom I, Gonzalez LC and Sheng M: TREM2 binds to apolipoproteins, including APOE and CLU/APOJ, and thereby facilitates uptake of Amyloid-beta by microglia. Neuron. 91:328–340. 2016. View Article : Google Scholar | |
Lessard CB, Malnik SL, Zhou Y, Ladd TB, Cruz PE, Ran Y, Mahan TE, Chakrabaty P, Holtzman DM, Ulrich JD, et al: High-affinity interactions and signal transduction between Aβ oligomers and TREM2. EMBO Mol Med. 10:e90272018. View Article : Google Scholar | |
Beisiegel U, Weber W and Bengtsson-Olivecrona G: Lipoprotein lipase enhances the binding of chylomicrons to low density lipoprotein receptor-related protein. Proc Natl Acad Sci USA. 88:8342–8346. 1991. View Article : Google Scholar | |
Gao Y, Vidal-Itriago A, Kalsbeek MJ, Layritz C, Garcia-Caceres C, Tom RZ, Eichmann TO, Vaz FM, Houtkooper RH, Van der Wel N, et al: Lipoprotein lipase maintains microglial innate immunity in obesity. Cell Rep. 20:3034–3042. 2017. View Article : Google Scholar : PubMed/NCBI | |
Aflaki E, Radovic B, Chandak PG, Kolb D, Eisenberg T, Ring J, Fertschai I, Uellen A, Wolinski H, Kohlwein SD, et al: Triacylglycerol accumulation activates the mitochondrial apoptosis pathway in macrophages. J Biol Chem. 286:7418–7428. 2011. View Article : Google Scholar : PubMed/NCBI | |
Weinger JG, Brosnan CF, Loudig O, Goldberg MF, Macian F, Arnett HA, Prieto AL, Tsiperson V and Shafit-Zagardo B: Loss of the receptor tyrosine kinase Axl leads to enhanced inflammation in the CNS and delayed removal of myelin debris during experimental autoimmune encephalomyelitis. J Neuroinflammation. 8:492011. View Article : Google Scholar : | |
Shirotani K, Hori Y, Yoshizaki R, Higuchi E, Colonna M, Saito T, Hashimoto S, Saito T, Saido TC and Iwata N: Aminophospholipids are Signal-transducing TREM2 ligands on apoptotic cells. Sci Rep. 9:75082019. View Article : Google Scholar : | |
Bogie JF, Jorissen W, Mailleux J, Nijland PG, Zelcer N, Vanmierlo T, Van Horssen J, Stinissen P, Hellings N and Hendriks JJ: Myelin alters the inflammatory phenotype of macrophages by activating PPARs. Acta Neuropathol Commun. 1:432013. View Article : Google Scholar | |
Bogie JF, Timmermans S, Huynh-Thu VA, Irrthum A, Smeets HJ, Gustafsson JA, Steffensen KR, Mulder M, Stinissen P, Hellings N and Hendriks JJ: Myelin-derived lipids modulate macrophage activity by liver X receptor activation. PLoS One. 7:e449982012. View Article : Google Scholar : PubMed/NCBI | |
Safaiyan S, Kannaiyan N, Snaidero N, Brioschi S, Biber K, Yona S, Edinger AL, Jung S, Rossner MJ and Simons M: Age-related myelin degradation burdens the clearance function of microglia during aging. Nat Neurosci. 19:995–998. 2016. View Article : Google Scholar | |
Gabande-Rodriguez E, Perez-Canamas A, Soto-Huelin B, Mitroi DN, Sanchez-Redondo S, Martinez-Saez E, Venero C, Peinado H and Ledesma MD: Lipid-induced lysosomal damage after demyelination corrupts microglia protective function in lysosomal storage disorders. EMBO J. 38:e995532019. View Article : Google Scholar | |
Carta AR and Simuni T: Thiazolidinediones under preclinical and early clinical development for the treatment of Parkinson's disease. Expert Opin Investig Drugs. 24:219–227. 2015. View Article : Google Scholar | |
Pioglitazone in early Parkinson's disease: A phase 2, multicentre, double-blind, randomised trial. Lancet Neurol. 14:795–803. 2015. View Article : Google Scholar | |
Huyghe S, Mannaerts GP, Baes M and Van Veldhoven PP: Peroxisomal multifunctional protein-2: The enzyme, the patients and the knockout mouse model. Biochim Biophys Acta. 1761:973–994. 2006. View Article : Google Scholar | |
Gong Y, Sasidharan N, Laheji F, Frosch M, Musolino P, Tanzi R, Kim DY, Biffi A, El Khoury J and Eichler F: Microglial dysfunction as a key pathological change in adrenomyeloneuropathy. Ann Neurol. 82:813–827. 2017. View Article : Google Scholar : PubMed/NCBI | |
Beckers L, Geric I, Stroobants S, Beel S, Van Damme P, D'Hooge R and Baes M: Microglia lacking a peroxisomal β-oxidation enzyme chronically alter their inflammatory profile without evoking neuronal and behavioral deficits. J Neuroinflammation. 16:612019. View Article : Google Scholar | |
Verheijden S, Beckers L, Casazza A, Butovsky O, Mazzone M and Baes M: Identification of a chronic non-neurodegenerative microglia activation state in a mouse model of peroxisomal β-oxidation deficiency. Glia. 63:1606–1620. 2015. View Article : Google Scholar | |
Sangineto M, Ciarnelli M, Cassano T, Radesco A, Moola A, Bukke VN, Romano A, Villani R, Kanwal H, Capitanio N, et al: Metabolic reprogramming in inflammatory microglia indicates a potential way of targeting inflammation in Alzheimer's disease. Redox Biol. 66:1028462023. View Article : Google Scholar : PubMed/NCBI | |
Meiser J, Kramer L, Sapcariu SC, Battello N, Ghelfi J, D'Herouel AF, Skupin A and Hiller K: Pro-inflammatory macrophages sustain pyruvate oxidation through pyruvate dehydrogenase for the synthesis of itaconate and to enable cytokine expression. J Biol Chem. 291:3932–3946. 2016. View Article : Google Scholar : | |
Lauterbach MA, Hanke JE, Serefidou M, Mangan MSJ, Kolbe CC, Hess T, Rothe M, Kaiser R, Hoss F, Gehlen J, et al: Toll-like receptor signaling rewires macrophage metabolism and promotes histone acetylation via ATP-citrate lyase. Immunity. 51:997–1011.e7. 2019. View Article : Google Scholar : PubMed/NCBI | |
Plastira I, Bernhart E, Joshi L, Koyani CN, Strohmaier H, Reicher H, Malle E and Sattler W: MAPK signaling determines lysophosphatidic acid (LPA)-induced inflammation in microglia. J Neuroinflammation. 17:1272020. View Article : Google Scholar : | |
Huang YL, Morales-Rosado J, Ray J, Myers TG, Kho T, Lu M and Munford RS: Toll-like receptor agonists promote prolonged triglyceride storage in macrophages. J Biol Chem. 289:3001–3012. 2014. View Article : Google Scholar : | |
Palmieri EM, Gonzalez-Cotto M, Baseler WA, Davies LC, Ghesquiere B, Maio N, Rice CM, Rouault TA, Cassel T, Higashi RM, et al: Nitric oxide orchestrates metabolic rewiring in M1 macrophages by targeting aconitase 2 and pyruvate dehydrogenase. Nat Commun. 11:6982020. View Article : Google Scholar | |
Rosas-Ballina M, Guan XL, Schmidt A and Bumann D: Classical activation of macrophages leads to lipid droplet formation without de novo fatty acid synthesis. Front Immunol. 11:1312020. View Article : Google Scholar : PubMed/NCBI | |
Bailey AP, Koster G, Guillermier C, Hirst EM, MacRae JI, Lechene CP, Postle AD and Gould AP: Antioxidant role for lipid droplets in a stem cell niche of drosophila. Cell. 163:340–353. 2015. View Article : Google Scholar : PubMed/NCBI | |
Khatchadourian A, Bourque SD, Richard VR, Titorenko VI and Maysinger D: Dynamics and regulation of lipid droplet formation in lipopolysaccharide (LPS)-stimulated microglia. Biochim Biophys Acta. 1821:607–617. 2012. View Article : Google Scholar : PubMed/NCBI | |
Marschallinger J, Iram T, Zardeneta M, Lee SE, Lehallier B, Haney MS, Pluvinage JV, Mathur V, Hahn O, Morgens DW, et al: Lipid-droplet-accumulating microglia represent a dysfunctional and proinflammatory state in the aging brain. Nat Neurosci. 23:194–208. 2020. View Article : Google Scholar : PubMed/NCBI | |
Bogie JFJ, Grajchen E, Wouters E, Corrales AG, Dierckx T, Vanherle S, Mailleux J, Gervois P, Wolfs E, Dehairs J, et al: Stearoyl-CoA desaturase-1 impairs the reparative properties of macrophages and microglia in the brain. J Exp Med. 217:e201916602020. View Article : Google Scholar : PubMed/NCBI | |
Szatmari I, Torocsik D, Agostini M, Nagy T, Gurnell M, Barta E, Chatterjee K and Nagy L: PPARgamma regulates the function of human dendritic cells primarily by altering lipid metabolism. Blood. 110:3271–3280. 2007. View Article : Google Scholar | |
Schonfeld P and Reiser G: Brain energy metabolism spurns fatty acids as fuel due to their inherent mitotoxicity and potential capacity to unleash neurodegeneration. Neurochem Int. 109:68–77. 2017. View Article : Google Scholar : PubMed/NCBI | |
Li LO, Klett EL and Coleman RA: Acyl-CoA synthesis, lipid metabolism and lipotoxicity. Biochim Biophys Acta. 1801:246–251. 2010. View Article : Google Scholar : | |
Fonteh AN, Cipolla M, Chiang J, Arakaki X and Harrington MG: Human cerebrospinal fluid fatty acid levels differ between supernatant fluid and brain-derived nanoparticle fractions, and are altered in Alzheimer's disease. PLoS One. 9:e1005192014. View Article : Google Scholar | |
Cunnane SC, Schneider JA, Tangney C, Tremblay-Mercier J, Fortier M, Bennett DA and Morris MC: Plasma and brain fatty acid profiles in mild cognitive impairment and Alzheimer's disease. J Alzheimers Dis. 29:691–697. 2012. View Article : Google Scholar | |
Snowden SG, Ebshiana AA, Hye A, An Y, Pletnikova O, O'Brien R, Troncoso J, Legido-Quigley C and Thambisetty M: Association between fatty acid metabolism in the brain and Alzheimer disease neuropathology and cognitive performance: A nontargeted metabolomic study. PLoS Med. 14:e10022662017. View Article : Google Scholar : | |
Belkouch M, Hachem M, Elgot A, Lo Van A, Picq M, Guichardant M, Lagarde M and Bernoud-Hubac N: The pleiotropic effects of omega-3 docosahexaenoic acid on the hallmarks of Alzheimer's disease. J Nutr Biochem. 38:1–11. 2016. View Article : Google Scholar | |
Fonteh AN, Cipolla M, Chiang AJ, Edminster SP, Arakaki X and Harrington MG: Polyunsaturated fatty acid composition of cerebrospinal fluid fractions shows their contribution to cognitive resilience of a Pre-symptomatic Alzheimer's disease cohort. Front Physiol. 11:832020. View Article : Google Scholar : PubMed/NCBI | |
Prasad MR, Lovell MA, Yatin M, Dhillon H and Markesbery WR: Regional membrane phospholipid alterations in Alzheimer's disease. Neurochem Res. 23:81–88. 1998. View Article : Google Scholar | |
Sebastiao AM, Colino-Oliveira M, Assaife-Lopes N, Dias RB and Ribeiro JA: Lipid rafts, synaptic transmission and plasticity: Impact in Age-related neurodegenerative diseases. Neuropharmacology. 64:97–107. 2013. View Article : Google Scholar | |
Sezgin E, Levental I, Mayor S and Eggeling C: The mystery of membrane organization: Composition, regulation and roles of lipid rafts. Nat Rev Mol Cell Biol. 18:361–374. 2017. View Article : Google Scholar : | |
Martin V, Fabelo N, Santpere G, Puig B, Marin R, Ferrer I and Diaz M: Lipid alterations in lipid rafts from Alzheimer's disease human brain cortex. J Alzheimers Dis. 19:489–502. 2010. View Article : Google Scholar | |
Fabelo N, Martin V, Marin R, Moreno D, Ferrer I and Diaz M: Altered lipid composition in cortical lipid rafts occurs at early stages of sporadic Alzheimer's disease and facilitates APP/BACE1 interactions. Neurobiol Aging. 35:1801–1812. 2014. View Article : Google Scholar | |
Filippov V, Song MA, Zhang K, Vinters HV, Tung S, Kirsch WM, Yang J and Duerksen-Hughes PJ: Increased ceramide in brains with Alzheimer's and other neurodegenerative diseases. J Alzheimers Dis. 29:537–547. 2012. View Article : Google Scholar | |
Cutler RG, Kelly J, Storie K, Pedersen WA, Tammara A, Hatanpaa K, Troncoso JC and Mattson MP: Involvement of oxidative Stress-induced abnormalities in ceramide and cholesterol metabolism in brain aging and Alzheimer's disease. Proc Natl Acad Sci USA. 101:2070–2075. 2004. View Article : Google Scholar : PubMed/NCBI | |
Han X, M Holtzman D, McKeel DW Jr, Kelley J and Morris JC: Substantial sulfatide deficiency and ceramide elevation in very early Alzheimer's disease: Potential role in disease pathogenesis. J Neurochem. 82:809–818. 2002. View Article : Google Scholar | |
He X, Huang Y, Li B, Gong CX and Schuchman EH: Deregulation of sphingolipid metabolism in Alzheimer's disease. Neurobiol Aging. 31:398–408. 2010. View Article : Google Scholar : | |
Soderberg M, Edlund C, Alafuzoff I, Kristensson K and Dallner G: Lipid composition in different regions of the brain in Alzheimer's disease/senile dementia of Alzheimer's type. J Neurochem. 59:1646–1653. 1992. View Article : Google Scholar : PubMed/NCBI | |
Siskind LJ: Mitochondrial ceramide and the induction of apoptosis. J Bioenerg Biomembr. 37:143–153. 2005. View Article : Google Scholar : PubMed/NCBI | |
Morris MC, Evans DA, Bienias JL, Tangney CC, Bennett DA, Wilson RS, Aggarwal N and Schneider J: Consumption of fish and n-3 fatty acids and risk of incident Alzheimer disease. Arch Neurol. 60:940–946. 2003. View Article : Google Scholar : PubMed/NCBI | |
Grimm MO, Grimm HS, Pätzold AJ, Zinser EG, Halonen R, Duering M, Tschape JA, De Strooper B, Müller U, Shen J and Hartmann T: Regulation of cholesterol and sphingomyelin metabolism by amyloid-beta and presenilin. Nat Cell Biol. 7:1118–1123. 2005. View Article : Google Scholar : PubMed/NCBI | |
Cheng H, Wang M, Li JL, Cairns NJ and Han X: Specific changes of sulfatide levels in individuals with pre-clinical Alzheimer's disease: An early event in disease pathogenesis. J Neurochem. 127:733–738. 2013. View Article : Google Scholar : PubMed/NCBI | |
Couttas TA, Kain N, Suchowerska AK, Quek LE, Turner N, Fath T, Garner B and Don AS: Loss of ceramide synthase 2 activity, necessary for myelin biosynthesis, precedes tau pathology in the cortical pathogenesis of Alzheimer's disease. Neurobiol Aging. 43:89–100. 2016. View Article : Google Scholar | |
Nasrabady SE, Rizvi B, Goldman JE and Brickman AM: White matter changes in Alzheimer's disease: A focus on myelin and oligodendrocytes. Acta Neuropathol Commun. 6:222018. View Article : Google Scholar : PubMed/NCBI | |
Heverin M, Bogdanovic N, Lutjohann D, Bayer T, Pikuleva I, Bretillon L, Diczfalusy U, Winblad B and Bjorkhem I: Changes in the levels of cerebral and extracerebral sterols in the brain of patients with Alzheimer's disease. J Lipid Res. 45:186–193. 2004. View Article : Google Scholar | |
Popp J, Meichsner S, Kolsch H, Lewczuk P, Maier W, Kornhuber J, Jessen F and Lutjohann D: Cerebral and extracerebral cholesterol metabolism and CSF markers of Alzheimer's disease. Biochem Pharmacol. 86:37–42. 2013. View Article : Google Scholar | |
Liu Y, Zhong X, Shen J, Jiao L, Tong J, Zhao W, Du K, Gong S, Liu M and Wei M: Elevated serum TC and LDL-C levels in Alzheimer's disease and mild cognitive impairment: A meta-analysis study. Brain Res. 1727:1465542020. View Article : Google Scholar | |
Zhang J and Liu Q: Cholesterol metabolism and homeostasis in the brain. Protein Cell. 6:254–264. 2015. View Article : Google Scholar : PubMed/NCBI | |
Tajima Y, Ishikawa M, Maekawa K, Murayama M, Senoo Y, Nishimaki-Mogami T, Nakanishi H, Ikeda K, Arita M and Taguchi R: Lipidomic analysis of brain tissues and plasma in a mouse model expressing mutated human amyloid precursor protein/tau for Alzheimer's disease. Lipids Health Dis. 12:682013. View Article : Google Scholar | |
Bhattacharyya R, Barren C and Kovacs DM: Palmitoylation of amyloid precursor protein regulates amyloidogenic processing in lipid rafts. J Neurosci. 33:11169–11183. 2013. View Article : Google Scholar : | |
Wang H, Kulas JA, Wang C, Holtzman DM, Ferris HA and Hansen SB: Regulation of beta-amyloid production in neurons by astrocyte-derived cholesterol. Proc Natl Acad Sci USA. 118:e21021911182021. View Article : Google Scholar | |
Qi G, Mi Y, Shi X, Gu H, Brinton RD and Yin F: ApoE4 impairs Neuron-astrocyte coupling of fatty acid metabolism. Cell Rep. 34:1085722021. View Article : Google Scholar : | |
Zhao J, Davis MD, Martens YA, Shinohara M, Graff-Radford NR, Younkin SG, Wszolek ZK, Kanekiyo T and Bu G: APOE ε4/ε4 diminishes neurotrophic function of human iPSC-derived astrocytes. Hum Mol Genet. 26:2690–2700. 2017. View Article : Google Scholar | |
Kober DL and Brett TJ: TREM2-ligand interactions in health and disease. J Mol Biol. 429:1607–1629. 2017. View Article : Google Scholar : PubMed/NCBI | |
Gouna G, Klose C, Bosch-Queralt M, Liu L, Gokce O, Schifferer M, Cantuti-Castelvetri L and Simons M: TREM2-dependent lipid droplet biogenesis in phagocytes is required for remyelination. J Exp Med. 218:e202102272021. View Article : Google Scholar : | |
Basil MC and Levy BD: Specialized Pro-resolving mediators: Endogenous regulators of infection and inflammation. Nat Rev Immunol. 16:51–67. 2016. View Article : Google Scholar | |
Whittington RA, Planel E and Terrando N: Impaired resolution of inflammation in Alzheimer's disease: A review. Front Immunol. 8:14642017. View Article : Google Scholar | |
Emre C, Hjorth E, Bharani K, Carroll S, Granholm AC and Schultzberg M: Receptors for Pro-resolving mediators are increased in Alzheimer's disease brain. Brain Pathol. 30:614–640. 2020. View Article : Google Scholar : PubMed/NCBI | |
Zhu M, Wang X, Hjorth E, Colas RA, Schroeder L, Granholm AC, Serhan CN and Schultzberg M: Pro-resolving lipid mediators improve neuronal survival and increase Aβ42 phagocytosis. Mol Neurobiol. 53:2733–2749. 2016. View Article : Google Scholar | |
Wang X, Zhu M, Hjorth E, Cortes-Toro V, Eyjolfsdottir H, Graff C, Nennesmo I, Palmblad J, Eriksdotter M, et al: Resolution of inflammation is altered in Alzheimer's disease. Alzheimers Dement. 11:40–50. e1–e2. 2015. View Article : Google Scholar | |
Malaplate-Armand C, Florent-Bechard S, Youssef I, Koziel V, Sponne I, Kriem B, Leininger-Muller B, Olivier JL, Oster T and Pillot T: Soluble oligomers of amyloid-beta peptide induce neuronal apoptosis by activating a cPLA2-dependent sphingomyelinase-ceramide pathway. Neurobiol Dis. 23:178–189. 2006. View Article : Google Scholar : PubMed/NCBI | |
Prasad VV, Nithipatikom K and Harder DR: Ceramide elevates 12-hydroxyeicosatetraenoic acid levels and upregulates 12-lipoxygenase in rat primary hippocampal cell cultures containing predominantly astrocytes. Neurochem Int. 53:220–229. 2008. View Article : Google Scholar | |
Assayag K, Yakunin E, Loeb V, Selkoe DJ and Sharon R: Polyunsaturated fatty acids induce alpha-synuclein-related pathogenic changes in neuronal cells. Am J Pathol. 171:2000–2011. 2007. View Article : Google Scholar | |
Flores-Leon M and Outeiro TF: More than meets the eye in Parkinson's disease and other synucleinopathies: From proteinopathy to lipidopathy. Acta Neuropathol. 146:369–385. 2023. View Article : Google Scholar | |
Yakunin E, Loeb V, Kisos H, Biala Y, Yehuda S, Yaari Y, Selkoe DJ and Sharon R: Α-synuclein neuropathology is controlled by nuclear hormone receptors and enhanced by docosahexaenoic acid in a mouse model for Parkinson's disease. Brain Pathol. 22:280–294. 2012. View Article : Google Scholar | |
Galvagnion C: The role of lipids interacting with α-synuclein in the pathogenesis of Parkinson's disease. J Parkinsons Dis. 7:433–450. 2017. View Article : Google Scholar | |
Garcia-Sanz P, MFG Aerts J and Moratalla R: The role of cholesterol in alpha-synuclein and lewy body pathology in GBA1 Parkinson's disease. Mov Disord. 36:1070–1085. 2021. View Article : Google Scholar | |
Paslawski W, Zareba-Paslawska J, Zhang X, Holzl K, Wadensten H, Shariatgorji M, Janelidze S, Hansson O, Forsgren L, Andrén PE and Svenningsson P: α-synuclein-lipoprotein interactions and elevated ApoE level in cerebrospinal fluid from Parkinson's disease patients. Proc Natl Acad Sci USA. 116:15226–15235. 2019. View Article : Google Scholar | |
Kim HE, Grant AR, Simic MS, Kohnz RA, Nomura DK, Durieux J, Riera CE, Sanchez M, Kapernick E, Wolff S and Dillin A: Lipid biosynthesis coordinates a mitochondrial-to-Cytosolic stress response. Cell. 166:1539–1552.e16. 2016. View Article : Google Scholar | |
Stein D, Mizrahi A, Golova A, Saretzky A, Venzor AG, Slobodnik Z, Kaluski S, Einav M, Khrameeva E and Toiber D: Aging and pathological aging signatures of the brain: Through the focusing lens of SIRT6. Aging (Albany NY). 13:6420–6441. 2021. View Article : Google Scholar | |
Smirnov D, Eremenko E, Stein D, Kaluski S, Jasinska W, Cosentino C, Martinez-Pastor B, Brotman Y, Mostoslavsky R, Khrameeva E and Toiber D: SIRT6 is a key regulator of mitochondrial function in the brain. Cell Death Dis. 14:352023. View Article : Google Scholar : PubMed/NCBI | |
Ghio S, Kamp F, Cauchi R, Giese A and Vassallo N: Interaction of α-synuclein with biomembranes in Parkinson's disease-role of cardiolipin. Prog Lipid Res. 61:73–82. 2016. View Article : Google Scholar | |
Chu CT, Ji J, Dagda RK, Jiang JF, Tyurina YY, Kapralov AA, Tyurin VA, Yanamala N, Shrivastava IH, Mohammadyani D, et al: Cardiolipin externalization to the outer mitochondrial membrane acts as an elimination signal for mitophagy in neuronal cells. Nat Cell Biol. 15:1197–1205. 2013. View Article : Google Scholar : PubMed/NCBI | |
Doblado L, Lueck C, Rey C, Samhan-Arias AK, Prieto I, Stacchiotti A and Monsalve M: Mitophagy in human diseases. Int J Mol Sci. 22:39032021. View Article : Google Scholar : | |
Rocha EM, Smith GA, Park E, Cao H, Graham AR, Brown E, McLean JR, Hayes MA, Beagan J, Izen SC, et al: Sustained systemic glucocerebrosidase inhibition induces Brain α-synuclein aggregation, microglia and complement C1q activation in mice. Antioxid Redox Signal. 23:550–564. 2015. View Article : Google Scholar : | |
Sampson TR, Debelius JW, Thron T, Janssen S, Shastri GG, Ilhan ZE, Challis C, Schretter CE, Rocha S, Gradinaru V, et al: Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson's disease. Cell. 167:1469–1480.e12. 2016. View Article : Google Scholar | |
Mogi M, Harada M, Kondo T, Riederer P, Inagaki H, Minami M and Nagatsu T: Interleukin-1 beta, interleukin-6, epidermal growth factor and transforming growth factor-alpha are elevated in the brain from parkinsonian patients. Neurosci Lett. 180:147–150. 1994. View Article : Google Scholar | |
Moloney EB, Moskites A, Ferrari EJ, Isacson O and Hallett PJ: The glycoprotein GPNMB is selectively elevated in the substantia nigra of Parkinson's disease patients and increases after lysosomal stress. Neurobiol Dis. 120:1–11. 2018. View Article : Google Scholar : PubMed/NCBI | |
Rebeck GW: The role of APOE on lipid homeostasis and inflammation in normal brains. J Lipid Res. 58:1493–1499. 2017. View Article : Google Scholar | |
Atagi Y, Liu CC, Painter MM, Chen XF, Verbeeck C, Zheng H, Li X, Rademakers R, Kang SS, Xu H, et al: Apolipoprotein E is a ligand for triggering receptor expressed on myeloid cells 2 (TREM2). J Biol Chem. 290:26043–26050. 2015. View Article : Google Scholar : PubMed/NCBI | |
Herrera AJ, Castano A, Venero JL, Cano J and Machado A: The single intranigral injection of LPS as a new model for studying the selective effects of inflammatory reactions on dopaminergic system. Neurobiol Dis. 7:429–447. 2000. View Article : Google Scholar | |
Gao HM, Kotzbauer PT, Uryu K, Leight S, Trojanowski JQ and Lee VM: Neuroinflammation and oxidation/nitration of alpha-synuclein linked to dopaminergic neurodegeneration. J Neurosci. 28:7687–7698. 2008. View Article : Google Scholar | |
Ioannou MS, Jackson J, Sheu SH, Chang CL, Weigel AV, Liu H, Pasolli HA, Xu CS, Pang S, Matthies D, et al: Neuron-astrocyte metabolic coupling protects against Activity-induced fatty acid toxicity. Cell. 177:1522–1535.e14. 2019. View Article : Google Scholar : PubMed/NCBI | |
Braidy N, Gai WP, Xu YH, Sachdev P, Guillemin GJ, Jiang XM, Ballard JW, Horan MP, Fang ZM, Chong BH and Chan DK: Uptake and mitochondrial dysfunction of alpha-synuclein in human astrocytes, cortical neurons and fibroblasts. Transl Neurodegener. 2:202013. View Article : Google Scholar : PubMed/NCBI | |
Sipione S, Rigamonti D, Valenza M, Zuccato C, Conti L, Pritchard J, Kooperberg C, Olson JM and Cattaneo E: Early transcriptional profiles in huntingtin-inducible striatal cells by microarray analyses. Hum Mol Genet. 11:1953–1965. 2002. View Article : Google Scholar : PubMed/NCBI | |
Leoni V and Caccia C: Study of cholesterol metabolism in Huntington's disease. Biochem Biophys Res Commun. 446:697–701. 2014. View Article : Google Scholar | |
Karasinska JM and Hayden MR: Cholesterol metabolism in Huntington disease. Nat Rev Neurol. 7:561–572. 2011. View Article : Google Scholar : PubMed/NCBI | |
Leoni V and Caccia C: The impairment of cholesterol metabolism in Huntington disease. Biochim Biophys Acta. 1851:1095–1105. 2015. View Article : Google Scholar | |
Valenza M, Rigamonti D, Goffredo D, Zuccato C, Fenu S, Jamot L, Strand A, Tarditi A, Woodman B, Racchi M, et al: Dysfunction of the cholesterol biosynthetic pathway in Huntington's disease. J Neurosci. 25:9932–9939. 2005. View Article : Google Scholar : PubMed/NCBI | |
Wang N, Yvan-Charvet L, Lutjohann D, Mulder M, Vanmierlo T, Kim TW and Tall AR: ATP-binding cassette transporters G1 and G4 mediate cholesterol and desmosterol efflux to HDL and regulate sterol accumulation in the brain. FASEB J. 22:1073–1082. 2008. View Article : Google Scholar | |
Leoni V, Long JD, Mills JA, Di Donato S and Paulsen JS; PREDICT-HD study group: Plasma 24S-hydroxycholesterol correlation with markers of Huntington disease progression. Neurobiol Dis. 55:37–43. 2013. View Article : Google Scholar : PubMed/NCBI | |
Tansey TR and Shechter I: Squalene synthase: Structure and regulation. Prog Nucleic Acid Res Mol Biol. 65:157–195. 2001. View Article : Google Scholar | |
Kreilaus F, Spiro AS, McLean CA, Garner B and Jenner AM: Evidence for altered cholesterol metabolism in Huntington's disease post mortem brain tissue. Neuropathol Appl Neurobiol. 42:535–546. 2016. View Article : Google Scholar | |
Valenza M, Leoni V, Tarditi A, Mariotti C, Bjorkhem I, Di Donato S and Cattaneo E: Progressive dysfunction of the cholesterol biosynthesis pathway in the R6/2 mouse model of Huntington's disease. Neurobiol Dis. 28:133–142. 2007. View Article : Google Scholar | |
Martin MG, Pfrieger F and Dotti CG: Cholesterol in brain disease: Sometimes determinant and frequently implicated. EMBO Rep. 15:1036–1052. 2014. View Article : Google Scholar : PubMed/NCBI | |
Valenza M, Leoni V, Karasinska JM, Petricca L, Fan J, Carroll J, Pouladi MA, Fossale E, Nguyen HP, Riess O, et al: Cholesterol defect is marked across multiple rodent models of Huntington's disease and is manifest in astrocytes. J Neurosci. 30:10844–10850. 2010. View Article : Google Scholar | |
Xu Z, He S, Begum MM and Han X: Myelin lipid alterations in neurodegenerative diseases: Landscape and pathogenic implications. Antioxid Redox Signal. 41:1073–1099. 2024. View Article : Google Scholar : PubMed/NCBI | |
Lawton KA, Brown MV, Alexander D, Li Z, Wulff JE, Lawson R, Jaffa M, Milburn MV, Ryals JA, Bowser R, et al: Plasma metabolomic biomarker panel to distinguish patients with amyotrophic lateral sclerosis from disease mimics. Amyotroph Lateral Scler Frontotemporal Degener. 15:362–370. 2014. View Article : Google Scholar | |
Goutman SA, Boss J, Guo K, Alakwaa FM, Patterson A, Kim S, Savelieff MG, Hur J and Feldman EL: Untargeted metabolomics yields insight into ALS disease mechanisms. J Neurol Neurosurg Psychiatry. 91:1329–1338. 2020. View Article : Google Scholar | |
Sol J, Jove M, Povedano M, Sproviero W, Dominguez R, Pinol-Ripoll G, Romero-Guevara R, Hye A, Al-Chalabi A, Torres P, et al: Lipidomic traits of plasma and cerebrospinal fluid in amyotrophic lateral sclerosis correlate with disease progression. Brain Commun. 3:fcab1432021. View Article : Google Scholar : PubMed/NCBI | |
Area-Gomez E, Larrea D, Yun T, Xu Y, Hupf J, Zandkarimi F, Chan RB and Mitsumoto H: Lipidomics study of plasma from patients suggest that ALS and PLS are part of a continuum of motor neuron disorders. Sci Rep. 11:135622021. View Article : Google Scholar | |
FernAndez-Eulate G, Ruiz-Sanz JI, Riancho J, ZufirIa M, GereNu G, FernAndez-TorrOn R, Poza-Aldea JJ, Ondaro J, Espinal JB, GonzÁlez-ChinchÓn G, et al: A comprehensive serum lipidome profiling of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener. 21:252–262. 2020. View Article : Google Scholar | |
Winkler EA, Sengillo JD, Sullivan JS, Henkel JS, Appel SH and Zlokovic BV: Blood-spinal cord barrier breakdown and pericyte reductions in amyotrophic lateral sclerosis. Acta Neuropathol. 125:111–120. 2013. View Article : Google Scholar | |
Waters S, Swanson MEV, Dieriks BV, Zhang YB, Grimsey NL, Murray HC, Turner C, Waldvogel HJ, Faull RLM, An J, et al: Blood-spinal cord barrier leakage is independent of motor neuron pathology in ALS. Acta Neuropathol Commun. 9:1442021. View Article : Google Scholar : PubMed/NCBI | |
Blasco H, Veyrat-Durebex C, Bocca C, Patin F, Vourc'h P, Kouassi Nzoughet J, Lenaers G, Andres CR, Simard G, Corcia P and Reynier P: Lipidomics reveals Cerebrospinal-Fluid signatures of ALS. Sci Rep. 7:176522017. View Article : Google Scholar | |
Patin F, Corcia P, Vourc'h P, Nadal-Desbarats L, Baranek T, Goossens JF, Marouillat S, Dessein AF, Descat A, Madji Hounoum B, et al: Omics to explore amyotrophic lateral sclerosis evolution: The central role of arginine and proline metabolism. Mol Neurobiol. 54:5361–5374. 2017. View Article : Google Scholar | |
Cutler RG, Pedersen WA, Camandola S, Rothstein JD and Mattson MP: Evidence that accumulation of ceramides and cholesterol esters mediates oxidative Stress-induced death of motor neurons in amyotrophic lateral sclerosis. Ann Neurol. 52:448–457. 2002. View Article : Google Scholar | |
Dodge JC, Jensen EH, Yu J, Sardi SP, Bialas AR, Taksir TV, Bangari DS and Shihabuddin LS: Neutral lipid cacostasis contributes to disease pathogenesis in amyotrophic lateral sclerosis. J Neurosci. 40:9137–9147. 2020. View Article : Google Scholar | |
Dodge JC, Treleaven CM, Pacheco J, Cooper S, Bao C, Abraham M, Cromwell M, Sardi SP, Chuang WL, Sidman RL, et al: Glycosphingolipids are modulators of disease pathogenesis in amyotrophic lateral sclerosis. Proc Natl Acad Sci USA. 112:8100–8105. 2015. View Article : Google Scholar : | |
Burg T, Rossaert E, Moisse M, Van Damme P and Van Den Bosch L: Histone deacetylase inhibition regulates lipid homeostasis in a mouse model of amyotrophic lateral sclerosis. Int J Mol Sci. 22:112242021. View Article : Google Scholar : | |
Chaves-Filho AB, Pinto IFD, Dantas LS, Xavier AM, Inague A, Faria RL, Medeiros MHG, Glezer I, Yoshinaga MY and Miyamoto S: Alterations in lipid metabolism of spinal cord linked to amyotrophic lateral sclerosis. Sci Rep. 9:116422019. View Article : Google Scholar : | |
Ramirez-Nunez O, Jove M, Torres P, Sol J, Fontdevila L, Romero-Guevara R, Andres-Benito P, Ayala V, Rossi C, Boada J, et al: Nuclear lipidome is altered in amyotrophic lateral sclerosis: A pilot study. J Neurochem. 158:482–499. 2021. View Article : Google Scholar | |
Johnson JO, Chia R, Miller DE, Li R, Kumaran R, Abramzon Y, Alahmady N, Renton AE, Topp SD, Gibbs JR, et al: Association of variants in the SPTLC1 gene with juvenile amyotrophic lateral sclerosis. JAMA Neurol. 78:1236–1248. 2021. View Article : Google Scholar | |
Mohassel P, Donkervoort S, Lone MA, Nalls M, Gable K, Gupta SD, Foley AR, Hu Y, Saute JAM, Moreira AL, et al: Childhood amyotrophic lateral sclerosis caused by excess sphingolipid synthesis. Nat Med. 27:1197–1204. 2021. View Article : Google Scholar | |
Kim SM, Noh MY, Kim H, Cheon SY, Lee KM, Lee J, Cha E, Park KS, Lee KW, Sung JJ and Kim SH: 25-Hydroxycholesterol is involved in the pathogenesis of amyotrophic lateral sclerosis. Oncotarget. 8:11855–11867. 2017. View Article : Google Scholar : PubMed/NCBI | |
Dodge JC, Yu J, Sardi SP and Shihabuddin LS: Sterol auto-oxidation adversely affects human motor neuron viability and is a neuropathological feature of amyotrophic lateral sclerosis. Sci Rep. 11:8032021. View Article : Google Scholar : PubMed/NCBI | |
Kann O: The interneuron energy hypothesis: Implications for brain disease. Neurobiol Dis. 90:75–85. 2016. View Article : Google Scholar | |
Mauch DH, Nägler K, Schumacher S, Göritz C, Müller EC, Otto A and Pfrieger FW: CNS synaptogenesis promoted by Glia-derived cholesterol. Science. 294:1354–1357. 2001. View Article : Google Scholar | |
Foley P: Lipids in Alzheimer's disease: A century-old story. Biochim Biophys Acta. 1801:750–753. 2010. View Article : Google Scholar | |
Cai XT, Li H, Borch Jensen M, Maksoud E, Borneo J, Liang Y, Quake SR, Luo L, Haghighi P and Jasper H: Gut cytokines modulate olfaction through metabolic reprogramming of glia. Nature. 596:97–102. 2021. View Article : Google Scholar : PubMed/NCBI | |
den Brok MH, Raaijmakers TK, Collado-Camps E and Adema GJ: Lipid droplets as immune modulators in myeloid cells. Trends Immunol. 39:380–392. 2018. View Article : Google Scholar | |
Lee JY, Marian OC and Don AS: Defective lysosomal lipid catabolism as a common pathogenic mechanism for dementia. Neuromolecular Med. 23:1–24. 2021. View Article : Google Scholar | |
Lin CH, Liao LY, Yang TY, Chang YJ, Tung CW, Hsu SL and Hsueh CM: Microglia-derived adiposomes are potential targets for the treatment of ischemic stroke. Cell Mol Neurobiol. 39:591–604. 2019. View Article : Google Scholar | |
Chali F, Milior G, Marty S, Morin-Brureau M, Le Duigou C, Savary E, Blugeon C, Jourdren L and Miles R: Lipid markers and related transcripts during excitotoxic neurodegeneration in kainate-treated mice. Eur J Neurosci. 50:1759–1778. 2019. View Article : Google Scholar | |
Astarita G, Jung KM, Vasilevko V, Dipatrizio NV, Martin SK, Cribbs DH, Head E, Cotman CW and Piomelli D: Elevated stearoyl-CoA desaturase in brains of patients with Alzheimer's disease. PLoS One. 6:e247772011. View Article : Google Scholar : PubMed/NCBI | |
Shibuya Y, Chang CC and Chang TY: ACAT1/SOAT1 as a therapeutic target for Alzheimer's disease. Future Med Chem. 7:2451–2467. 2015. View Article : Google Scholar | |
Lin YT, Seo J, Gao F, Feldman HM, Wen HL, Penney J, Cam HP, Gjoneska E, Raja WK, Cheng J, et al: APOE4 causes widespread molecular and cellular alterations associated with Alzheimer's disease phenotypes in human iPSC-derived brain cell types. Neuron. 98:1141–1154.e7. 2018. View Article : Google Scholar | |
Chen Y, Strickland MR, Soranno A and Holtzman DM: Apolipoprotein E: Structural Insights and Links to Alzheimer disease pathogenesis. Neuron. 109:205–221. 2021. View Article : Google Scholar | |
Yen JHJ and Yu ICI: The role of ApoE-mediated microglial lipid metabolism in brain aging and disease. Immunometabolism (Cobham). 5:e000182023. View Article : Google Scholar | |
Sienski G, Narayan P, Bonner JM, Kory N, Boland S, Arczewska AA, Ralvenius WT, Akay L, Lockshin E, He L, et al: APOE4 disrupts intracellular lipid homeostasis in human iPSC-derived glia. Sci Transl Med. 13:eaaz45642021. View Article : Google Scholar : PubMed/NCBI | |
Tcw J, Qian L, Pipalia NH, Chao MJ, Liang SA, Shi Y, Jain BR, Bertelsen SE, Kapoor M, Marcora E, et al: Cholesterol and matrisome pathways dysregulated in astrocytes and microglia. Cell. 185:2213–2233.e25. 2022. View Article : Google Scholar : PubMed/NCBI | |
Victor MB, Leary N, Luna X, Meharena HS, Scannail AN, Bozzelli PL, Samaan G, Murdock MH, von Maydell D, Effenberger AH, et al: Lipid accumulation induced by APOE4 impairs microglial surveillance of neuronal-network activity. Cell Stem Cell. 29:1197–1212.e8. 2022. View Article : Google Scholar : PubMed/NCBI | |
Guglielmotto M, Monteleone D, Piras A, Valsecchi V, Tropiano M, Ariano S, Fornaro M, Vercelli A, Puyal J, Arancio O, et al: Aβ1-42 monomers or oligomers have different effects on autophagy and apoptosis. Autophagy. 10:1827–1843. 2014. View Article : Google Scholar : PubMed/NCBI | |
Baerends E, Soud K, Folke J, Pedersen AK, Henmar S, Konrad L, Lycas MD, Mori Y, Pakkenberg B, Woldbye DPD, et al: Modeling the early stages of Alzheimer's disease by administering intracerebroventricular injections of human native Aβ oligomers to rats. Acta Neuropathol Commun. 10:1132022. View Article : Google Scholar | |
Brown AJ and Jessup W: Oxysterols: Sources, cellular storage and metabolism, and new insights into their roles in cholesterol homeostasis. Mol Aspects Med. 30:111–122. 2009. View Article : Google Scholar | |
Chang JY, Chavis JA, Liu LZ and Drew PD: Cholesterol oxides induce programmed cell death in microglial cells. Biochem Biophys Res Commun. 249:817–821. 1998. View Article : Google Scholar | |
Liu J, Liu Y, Chen J, Hu C, Teng M, Jiao K, Shen Z, Zhu D, Yue J, Li Z and Li Y: The ROS-mediated activation of IL-6/STAT3 signaling pathway is involved in the 27-hydroxycholesterol-induced cellular senescence in nerve cells. Toxicol In Vitro. 45:10–18. 2017. View Article : Google Scholar : PubMed/NCBI | |
Simpson DSA and Oliver PL: ROS Generation in microglia: Understanding oxidative stress and inflammation in neurodegenerative disease. Antioxidants (Basel). 9:7432020. View Article : Google Scholar : PubMed/NCBI | |
Olsen BN, Schlesinger PH and Baker NA: Perturbations of membrane structure by cholesterol and cholesterol derivatives are determined by sterol orientation. J Am Chem Soc. 131:4854–4865. 2009. View Article : Google Scholar : PubMed/NCBI | |
Kauffman JM, Westerman PW and Carey MC: Fluorocholesterols, in contrast to hydroxycholesterols, exhibit interfacial properties similar to cholesterol. J Lipid Res. 41:991–1003. 2000. View Article : Google Scholar : PubMed/NCBI | |
Appelqvist H, Wäster P, Kågedal K and Öllinger K: The lysosome: From waste bag to potential therapeutic target. J Mol Cell Biol. 5:214–226. 2013. View Article : Google Scholar : PubMed/NCBI | |
Gosselet F, Saint-Pol J and Fenart L: Effects of oxysterols on the blood-brain barrier: Implications for Alzheimer's disease. Biochem Biophys Res Commun. 446:687–691. 2014. View Article : Google Scholar | |
Trompier D, Vejux A, Zarrouk A, Gondcaille C, Geillon F, Nury T, Savary S and Lizard G: Brain peroxisomes. Biochimie. 98:102–110. 2014. View Article : Google Scholar | |
Loving BA, Tang M, Neal MC, Gorkhali S, Murphy R, Eckel RH and Bruce KD: Lipoprotein lipase regulates microglial lipid droplet accumulation. Cells. 10:1982021. View Article : Google Scholar | |
Berghoff SA, Spieth L, Sun T, Hosang L, Schlaphoff L, Depp C, Düking T, Winchenbach J, Neuber J, Ewers D, et al: Microglia facilitate repair of demyelinated lesions via Post-squalene sterol synthesis. Nat Neurosci. 24:47–60. 2021. View Article : Google Scholar : | |
Ciesielska A, Matyjek M and Kwiatkowska K: TLR4 and CD14 trafficking and its influence on LPS-induced Pro-inflammatory signaling. Cell Mol Life Sci. 78:1233–1261. 2021. View Article : Google Scholar | |
Sheng JG, Bora SH, Xu G, Borchelt DR, Price DL and Koliatsos VE: Lipopolysaccharide-Induced-neuroinflammation increases intracellular accumulation of amyloid precursor protein and amyloid beta peptide in APPswe transgenic mice. Neurobiol Dis. 14:133–145. 2003. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Yin M, Cao X, Hu G and Xiao M: Pro- and Anti-inflammatory effects of high cholesterol diet on aged brain. Aging Dis. 9:374–390. 2018. View Article : Google Scholar : PubMed/NCBI | |
Iannucci J, Sen A and Grammas P: Isoform-specific effects of apolipoprotein E on markers of inflammation and toxicity in brain glia and neuronal cells in vitro. Curr Issues Mol Biol. 43:215–225. 2021. View Article : Google Scholar | |
Churchward MA and Todd KG: Statin treatment affects cytokine release and phagocytic activity in primary cultured microglia through two separable mechanisms. Mol Brain. 7:852014. View Article : Google Scholar : PubMed/NCBI | |
Tanaka N, Abe-Dohmae S, Iwamoto N, Fitzgerald ML and Yokoyama S: Helical apolipoproteins of High-density lipoprotein enhance phagocytosis by stabilizing ATP-binding cassette transporter A7. J Lipid Res. 51:2591–2599. 2010. View Article : Google Scholar : PubMed/NCBI | |
Aikawa T, Holm ML and Kanekiyo T: ABCA7 and pathogenic pathways of Alzheimer's disease. Brain Sci. 8:272018. View Article : Google Scholar : PubMed/NCBI | |
Dai W, Yao RM, Mi TY, Zhang LM, Wu HL, Cheng JB and Li YF: Cognition-enhancing effect of YL-IPA08, a potent ligand for the translocator protein (18 kDa) in the 5 x FAD transgenic mouse model of Alzheimer's pathology. J Psychopharmacol. 36:1176–1187. 2022. View Article : Google Scholar : PubMed/NCBI | |
Bouhrara M, Reiter DA, Bergeron CM, Zukley LM, Ferrucci L, Resnick SM and Spencer RG: Evidence of demyelination in mild cognitive impairment and dementia using a direct and specific magnetic resonance imaging measure of myelin content. Alzheimers Dement. 14:998–1004. 2018. View Article : Google Scholar | |
Benitez A, Fieremans E, Jensen JH, Falangola MF, Tabesh A, Ferris SH and Helpern JA: White matter tract integrity metrics reflect the vulnerability of Late-myelinating tracts in Alzheimer's disease. Neuroimage Clin. 4:64–71. 2014. View Article : Google Scholar | |
Depp C, Sun T, Sasmita AO, Spieth L, Berghoff SA, Nazarenko T, Overhoff K, Steixner-Kumar AA, Subramanian S, Arinrad S, et al: Myelin dysfunction drives amyloid-β deposition in models of Alzheimer's disease. Nature. 618:349–357. 2023. View Article : Google Scholar : PubMed/NCBI | |
Mathys H, Davila-Velderrain J, Peng Z, Gao F, Mohammadi S, Young JZ, Menon M, He L, Abdurrob F, Jiang X, et al: Single-cell transcriptomic analysis of Alzheimer's disease. Nature. 570:332–337. 2019. View Article : Google Scholar : PubMed/NCBI | |
Saez-Atienzar S and Masliah E: Cellular senescence and Alzheimer disease: The egg and the chicken scenario. Nat Rev Neurosci. 21:433–444. 2020. View Article : Google Scholar : PubMed/NCBI | |
Lloyd AF and Miron VE: The Pro-remyelination properties of microglia in the central nervous system. Nat Rev Neurol. 15:447–458. 2019. View Article : Google Scholar | |
Cignarella F, Filipello F, Bollman B, Cantoni C, Locca A, Mikesell R, Manis M, Ibrahim A, Deng L, Benitez BA, et al: TREM2 activation on microglia promotes myelin debris clearance and remyelination in a model of multiple sclerosis. Acta Neuropathol. 140:513–534. 2020. View Article : Google Scholar : PubMed/NCBI | |
Miron VE, Boyd A, Zhao JW, Yuen TJ, Ruckh JM, Shadrach JL, van Wijngaarden P, Wagers AJ, Williams A, Franklin RJM, et al: M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat Neurosci. 16:1211–1218. 2013. View Article : Google Scholar : PubMed/NCBI | |
Lloyd AF, Davies CL, Holloway RK, Labrak Y, Ireland G, Carradori D, Dillenburg A, Borger E, Soong D, Richardson JC, et al: Central nervous system regeneration is driven by microglia necroptosis and repopulation. Nat Neurosci. 22:1046–1052. 2019. View Article : Google Scholar : | |
Mecha M, Yanguas-Casás N, Feliú A, Mestre L, Carrillo-Salinas F, Azcoitia I, Yong VW and Guaza C: The endocannabinoid 2-AG enhances spontaneous remyelination by targeting microglia. Brain Behav Immun. 77:110–126. 2019. View Article : Google Scholar | |
Wang X, Cao K, Sun X, Chen Y, Duan Z, Sun L, Guo L, Bai P, Sun D, Fan J, et al: Macrophages in spinal cord injury: Phenotypic and functional change from exposure to myelin debris. Glia. 63:635–651. 2015. View Article : Google Scholar : | |
Mrdjen D, Pavlovic A, Hartmann FJ, Schreiner B, Utz SG, Leung BP, Lelios I, Heppner FL, Kipnis J, Merkler D, et al: High-Dimensional Single-cell mapping of central nervous system immune cells reveals distinct myeloid subsets in health, aging, and disease. Immunity. 48:5992018. View Article : Google Scholar | |
Hammond TR, Dufort C, Dissing-Olesen L, Giera S, Young A, Wysoker A, Walker AJ, Gergits F, Segel M, Nemesh J, et al: Single-Cell RNA sequencing of microglia throughout the mouse lifespan and in the injured brain reveals complex cell-state changes. Immunity. 50:253–271.e6. 2019. View Article : Google Scholar | |
Zrzavy T, Hametner S, Wimmer I, Butovsky O, Weiner HL and Lassmann H: Loss of 'homeostatic' microglia and patterns of their activation in active multiple sclerosis. Brain. 140:1900–1913. 2017. View Article : Google Scholar : PubMed/NCBI | |
Locatelli G, Theodorou D, Kendirli A, Jordão MJC, Staszewski O, Phulphagar K, Cantuti-Castelvetri L, Dagkalis A, Bessis A, Simons M, et al: Mononuclear phagocytes locally specify and adapt their phenotype in a multiple sclerosis model. Nat Neurosci. 21:1196–1208. 2018. View Article : Google Scholar : PubMed/NCBI | |
Wlodarczyk A, Benmamar-Badel A, Cédile O, Jensen KN, Kramer I, Elsborg NB and Owens T: CSF1R stimulation promotes increased neuroprotection by CD11c+ microglia in EAE. Front Cell Neurosci. 12:5232018. View Article : Google Scholar | |
Yu Z, Sun D, Feng J, Tan W, Fang X, Zhao M, Zhao X, Pu Y, Huang A, Xiang Z, et al: MSX3 Switches microglia polarization and protects from Inflammation-induced demyelination. J Neurosci. 35:6350–6365. 2015. View Article : Google Scholar | |
Zabala A, Vazquez-Villoldo N, Rissiek B, Gejo J, Martin A, Palomino A, Perez-Samartín A, Pulagam KR, Lukowiak M, Capetillo-Zarate E, et al: P2X4 receptor controls microglia activation and favors remyelination in autoimmune encephalitis. EMBO Mol Med. 10:e87432018. View Article : Google Scholar : | |
Brüne B, Dehne N, Grossmann N, Jung M, Namgaladze D, Schmid T, von Knethen A and Weigert A: Redox control of inflammation in macrophages. Antioxid Redox Signal. 19:595–637. 2013. View Article : Google Scholar | |
Conrad M, Kagan VE, Bayir H, Pagnussat GC, Head B, Traber MG and Stockwell BR: Regulation of lipid peroxidation and ferroptosis in diverse species. Genes Dev. 32:602–619. 2018. View Article : Google Scholar : PubMed/NCBI | |
Wenzel SE, Tyurina YY, Zhao J, St Croix CM, Dar HH, Mao G, Tyurin VA, Anthonymuthu TS, Kapralov AA, Amoscato AA, et al: PEBP1 wardens ferroptosis by enabling lipoxygenase generation of lipid death signals. Cell. 171:628–641.e26. 2017. View Article : Google Scholar : PubMed/NCBI | |
Kagan VE, Mao G, Qu F, Angeli JP, Doll S, Croix CS, Dar HH, Liu B, Tyurin VA, Ritov VB, et al: Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol. 13:81–90. 2017. View Article : Google Scholar | |
Anthonymuthu TS, Kenny EM, Shrivastava I, Tyurina YY, Hier ZE, Ting HC, Dar HH, Tyurin VA, Nesterova A, Amoscato AA, et al: Empowerment of 15-lipoxygenase catalytic competence in selective oxidation of membrane ETE-PE to ferroptotic death signals, HpETE-PE. J Am Chem Soc. 140:17835–17839. 2018. View Article : Google Scholar : PubMed/NCBI | |
Kapralov AA, Yang Q, Dar HH, Tyurina YY, Anthonymuthu TS, Kim R, St Croix CM, Mikulska-Ruminska K, Liu B, Shrivastava IH, et al: Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death. Nat Chem Biol. 16:278–290. 2020. View Article : Google Scholar : PubMed/NCBI | |
Doll S, Proneth B, Tyurina YY, Panzilius E, Kobayashi S, Ingold I, Irmler M, Beckers J, Aichler M, Walch A, et al: ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 13:91–98. 2017. View Article : Google Scholar | |
Zhou Y, Yang Y, Yi L, Pan M, Tang W and Duan H: Propofol mitigates Sepsis-induced brain injury by inhibiting ferroptosis via activation of the Nrf2/HO-1axis. Neurochem Res. 49:2131–2147. 2024. View Article : Google Scholar | |
Zhou X, Zhao R, Lv M, Xu X, Liu W, Li X, Gao Y, Zhao Z, Zhang Z, Li Y, et al: ACSL4 promotes microgliamediated neuroinflammation by regulating lipid metabolism and VGLL4 expression. Brain Behav Immun. 109:331–343. 2023. View Article : Google Scholar : PubMed/NCBI | |
Liang D, Minikes AM and Jiang X: Ferroptosis at the intersection of lipid metabolism and cellular signaling. Mol Cell. 82:2215–2227. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wu H, Li N, Peng S, Fu H, Hu Z and Su L: Maresin1 improves hippocampal neuroinflammation and cognitive function in septic rats by activating the SLC7A11/GPX4 ferroptosis signaling pathway. Int Immunopharmacol. 131:1117922024. View Article : Google Scholar | |
Fernández-Mendívil C, Luengo E, Trigo-Alonso P, García-Magro N, Negredo P and López MG: Protective role of microglial HO-1 blockade in aging: Implication of iron metabolism. Redox Biol. 38:1017892021. View Article : Google Scholar | |
Gao S, Zhou L, Lu J, Fang Y, Wu H, Xu W, Pan Y, Wang J, Wang X, Zhang J and Shao A: Cepharanthine attenuates early brain injury after subarachnoid hemorrhage in mice via inhibiting 15-Lipoxygenase-1-Mediated microglia and endothelial cell ferroptosis. Oxid Med Cell Longev. 2022:42952082022. View Article : Google Scholar : | |
Strike SC, Carlisle A, Gibson EL and Dyall SC: A High Omega-3 fatty acid multinutrient supplement benefits cognition and mobility in older women: A randomized, Double-blind, Placebo-controlled pilot study. J Gerontol A Biol Sci Med Sci. 71:236–242. 2016. View Article : Google Scholar | |
Lee LK, Shahar S, Chin AV and Yusoff NA: Docosahexaenoic Acid-concentrated fish oil supplementation in subjects with mild cognitive impairment (MCI): A 12-month randomised, double-blind, placebo-controlled trial. Psychopharmacology (Berl). 225:605–612. 2013. View Article : Google Scholar | |
Serrano-Pozo A, Vega GL, Lütjohann D, Locascio JJ, Tennis MK, Deng A, Atri A, Hyman BT, Irizarry MC and Growdon JH: Effects of simvastatin on cholesterol metabolism and Alzheimer disease biomarkers. Alzheimer Dis Assoc Disord. 24:220–226. 2010. View Article : Google Scholar | |
Evans BA, Evans JE, Baker SP, Kane K, Swearer J, Hinerfeld D, Caselli R, Rogaeva E, St George-Hyslop P, Moonis M and Pollen DA: Long-term statin therapy and CSF cholesterol levels: Implications for Alzheimer's disease. Dement Geriatr Cogn Disord. 27:519–524. 2009. View Article : Google Scholar : PubMed/NCBI | |
Qin Z, Gu M, Zhou J, Zhang W, Zhao N, Lü Y and Yu W: Triggering receptor expressed on myeloid cells 2 activation downregulates toll-like receptor 4 expression and ameliorates cognitive impairment in the Aβ1-42-induced Alzheimer's disease mouse model. Synapse. 74:e221612020. View Article : Google Scholar | |
Price BR, Sudduth TL, Weekman EM, Johnson S, Hawthorne D, Woolums A and Wilcock DM: Therapeutic Trem2 activation ameliorates amyloid-beta deposition and improves cognition in the 5XFAD model of amyloid deposition. J Neuroinflammation. 17:2382020. View Article : Google Scholar : PubMed/NCBI | |
Fitz NF, Nam KN, Wolfe CM, Letronne F, Playso BE, Iordanova BE, Kozai TDY, Biedrzycki RJ, Kagan VE, Tyurina YY, et al: Phospholipids of APOE lipoproteins activate microglia in an Isoform-specific manner in preclinical models of Alzheimer's disease. Nat Commun. 12:34162021. View Article : Google Scholar : PubMed/NCBI | |
Griciuc A, Patel S, Federico AN, Choi SH, Innes BJ, Oram MK, Cereghetti G, McGinty D, Anselmo A, Sadreyev RI, et al: TREM2 Acts downstream of CD33 in modulating microglial pathology in Alzheimer's disease. Neuron. 103:820–835.e827. 2019. View Article : Google Scholar | |
Lefterov I, Schug J, Mounier A, Nam KN, Fitz NF and Koldamova R: RNA-sequencing reveals transcriptional up-regulation of Trem2 in response to bexarotene treatment. Neurobiol Dis. 82:132–140. 2015. View Article : Google Scholar : PubMed/NCBI | |
Tai LM, Koster KP, Luo J, Lee SH, Wang YT, Collins NC, Ben Aissa M, Thatcher GRJ and LaDu MJ: Amyloid-β pathology and APOE genotype modulate retinoid X receptor agonist activity in vivo. J Biol Chem. 289:30538–30555. 2014. View Article : Google Scholar : | |
Khan N, Syed DN, Ahmad N and Mukhtar H: Fisetin: A dietary antioxidant for health promotion. Antioxid Redox Signal. 19:151–162. 2013. View Article : Google Scholar : | |
Prior M, Chiruta C, Currais A, Goldberg J, Ramsey J, Dargusch R, Maher PA and Schubert D: Back to the future with phenotypic screening. ACS Chem Neurosci. 5:503–513. 2014. View Article : Google Scholar | |
Ates G, Goldberg J, Currais A and Maher P: CMS121, a fatty acid synthase inhibitor, protects against excess lipid peroxidation and inflammation and alleviates cognitive loss in a transgenic mouse model of Alzheimer's disease. Redox Biol. 36:1016482020. View Article : Google Scholar : PubMed/NCBI | |
Ayala A, Muñoz MF and Argüelles S: Lipid peroxidation: Production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid Med Cell Longev. 2014:3604382014. View Article : Google Scholar : PubMed/NCBI | |
Oostveen JA, Dunn E, Carter DB and Hall ED: Neuroprotective efficacy and mechanisms of novel pyrrolopyrimidine lipid peroxidation inhibitors in the gerbil forebrain ischemia model. J Cereb Blood Flow Metab. 18:539–547. 1998. View Article : Google Scholar |