Next Article in Journal
Biomarkers and Breakdowns: Neuroinflammatory Drivers Linking Sleep Disorders and Chronic Pain
Previous Article in Journal
Editorial: Genomic Insights and Translational Opportunities for Human Cancers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Glial Cell Dynamics in Neuroinflammation: Mechanisms, Interactions, and Therapeutic Implications

by
Mario García-Domínguez
1,2,3
1
Program of Immunology and Immunotherapy, CIMA-Universidad de Navarra, 31008 Pamplona, Spain
2
Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
3
Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
Biomedicines 2026, 14(1), 115; https://doi.org/10.3390/biomedicines14010115
Submission received: 29 November 2025 / Revised: 23 December 2025 / Accepted: 4 January 2026 / Published: 6 January 2026

Abstract

Neuroinflammation is a defining feature of many neurological disorders, including neurodegenerative diseases, traumatic brain injury, and demyelinating conditions. Glial cells play a central role in this process by initiating, modulating, and resolving inflammatory responses in the CNS. This review examines the diverse roles of glial cells in neuroinflammation, focusing on their molecular and cellular interactions, context-dependent activation states, and phenotypic plasticity. It discusses how microglial activation can result in both neuroprotective and neurotoxic effects, while astrocytes contribute to immune regulation, blood–brain barrier integrity, and neuronal survival. The review also highlights interactions between glial cells and peripheral immune components, which may exert synergistic or antagonistic effects. Finally, it outlines emerging preclinical and clinical strategies targeting glial pathways to modulate several neuroinflammatory outcomes, emphasizing that a detailed understanding of glial dynamics is essential for developing effective CNS therapies.

1. Introduction

Neuroinflammation is a complex process that is involved in numerous diseases of the nervous system [1,2], which include acute injuries such as traumatic brain injury (TBI) [3] and stroke [4], as well as chronic neurodegenerative disorders like Alzheimer’s disease (AD) [5], Parkinson’s disease (PD) [6], multiple sclerosis (MS) [7], and amyotrophic lateral sclerosis (ALS) [8]. In contrast to peripheral inflammation, neuroinflammation is coordinated in the microenvironment of the central nervous system (CNS), in which specialized immune-competent cells, known as glial cells, play a pivotal regulatory role [9]. Glial cells which include microglial cells, astrocytes, oligodendrocytes, and their progenitors, are not only the primary mediators of the CNS innate immune response but also the main contributors to the homeostasis, synaptic remodeling, and neuronal support under physiological conditions [10,11].
Upon exposure to pathological stimuli or injury, glial cells undergo phenotypic and functional modifications, a phenomenon broadly referred to as glial activation [12]. This dynamic response encompasses coordinated changes in morphology, transcriptional and translational circuits, secretory functions, and intercellular communication with neurons as well as other glial cell types [13]. Glial activation serves as a defining characteristic of the CNS’s response to a diverse array of insults, such as trauma, infection, ischemia, and neurodegenerative processes [14].
The first line of defense against damage or pathogen invasion is provided by microglial cells, the CNS’s resident innate immune cells and main phagocytes [15]. Due to their high degree of plasticity, microglial cells can quickly switch between two activation states. Traditionally, these have ranged from pro-inflammatory phenotype (M1-like) that can exacerbate neuronal damage to anti-inflammatory (M2-like) that aid in tissue regeneration and repair [16]. Numerous environmental cues, including cytokines, pathogen-associated molecular patterns, and damage-associated molecular patterns (DAMPs), influence this spectrum of activation [17].
The most predominant glial cell type, astrocytes, are also fundamental for maintaining homeostasis in the CNS and responding to injury [18]. When astrocytes are activated, they exhibit reactive astrogliosis, which is typified by changes in gene expression profiles, hypertrophy, and upregulation of intermediate filaments like glial fibrillary acidic protein (GFAP) [19]. Reactive astrocytes have many functions, including regulating extracellular ion and neurotransmitter concentrations, secreting numerous pro- and anti-inflammatory mediators, influencing synaptic plasticity, and influencing neuronal metabolism and permeability of the blood–brain barrier (BBB) [20]. Beyond their crucial role in mediating immune signaling within the CNS, oligodendrocytes are commonly known for their function in the development and upkeep of myelin sheaths surrounding axons [21]. By releasing a variety of cytokines, oligodendrocytes can contribute to neuroinflammation under pathological conditions. They might also participate in remyelination and axonal support processes [22]. Furthermore, compared to earlier estimates, their interactions with other glial cells point to a more integrated role in the larger glial response to CNS insult [23].
A dynamic and complex network of interactions, essential for the initiation and resolution of CNS immune responses, is exemplified by the interplay among glial subtypes during neuroinflammation [24]. The release and reception of cytokines, extracellular vesicles (such as exosomes and microvesicles), and direct physical interactions through specific cell–cell contact sites are all part of this bidirectional communication, which involves a complex interplay of signaling pathways [25]. In this neuroimmune network, each type of glial cell shapes both local and systemic responses to injury or disease through unique but interdependent roles [26].
In order to describe the role of glial cells in the development, progression, and resolution of neuropathological conditions, the idea of disease-associated glial cells has gained popularity [27]. Molecularly distinct glial subpopulations that arise in response to disease-specific perturbations, such as demyelination in MD or protein aggregation in other neurodegenerative disorders, have been identified thanks to high-definition techniques like scRNA-seq (single-cell RNA sequencing) and spatial transcriptomics [28,29]. These transcriptionally altered states are characterized by changes in immune signaling, metabolic pathways, synaptic modulation, and phagocytic activity, and they represent context-dependent adaptations rather than just reactive phenotypes [30]. Specifically, single-cell RNA sequencing (scRNA-seq) studies have revealed that the classical M1/M2 framework is overly simplistic and fails to accurately represent the full transcriptional and functional diversity of microglial cells. Rather than existing as binary activation states, microglia display a dynamic continuum of context-dependent phenotypes that are highly sensitive to developmental stage, aging, regional CNS environment, and pathological insults [31,32]. These include homeostatic microglia that maintain synaptic pruning and tissue surveillance, developmental- and aging-associated subpopulations with stage-specific transcriptional programs, disease-associated microglia (DAM) that emerge in neurodegenerative conditions, and injury-induced phenotypes that respond to tissue damage. Beyond these broad categories, scRNA-seq analyses have further identified microglial subsets with specialized functions, including antigen-presenting cells capable of modulating adaptive immune responses, interferon-responsive populations that participate in antiviral defense, metabolically distinct microglia optimized for energy-intensive processes, and highly proliferative subsets involved in local tissue repopulation. Importantly, these subpopulations exhibit regional specialization within the CNS, reflecting the unique microenvironmental cues of different brain and spinal cord regions [31,32].
Analogously, astrocytes also exhibit remarkable heterogeneity in response to pathological stimuli. Under disease, scRNA-seq studies have identified multiple astrocytic subtypes defined by distinct metabolic pathways, differential expression of neurotrophic and synaptogenic factors, and specialized roles in modulating neuronal activity, synaptic remodeling, and blood–brain barrier integrity [33]. Certain astrocyte populations contribute to protective processes, such as supporting neuronal survival and facilitating synaptic recovery, whereas others may adopt maladaptive profiles that exacerbate inflammation or synaptic dysfunction [33].
Glial cell heterogeneity, plasticity, and context-dependent functions are now understood thanks to many developments in molecular and imaging technologies, which have also revealed intricate regulatory networks that control how these cells behave in health and illness. As a result, glial cell targeting offers an effective therapeutic approach to reduce neuroinflammatory responses and encourage neuroprotection and repair [34]. Current research focusses on pharmacological approaches, such as gene-targeting strategies, biologics, and small-molecule modulators, to modify glial activation, reduce neuroinflammation, and support neuroprotection and repair [35,36,37,38,39].
This review provides a focused analysis of recent discoveries on the temporal and spatial dynamics of glial cell activation and their interactions during neuroinflammation. Centered on the question of how these dynamics shape CNS inflammatory responses and impact disease progression, it emphasizes mechanistic pathways through which glial cells detect, propagate, and resolve neuroinflammatory signals, highlighting those most relevant for translational and therapeutic applications. Rather than attempting a comprehensive catalog of all facets of glial biology, this review emphasizes mechanisms with demonstrated functional significance and translational relevance, while more speculative or less-established pathways are noted as directions for future research. By integrating evidence from cellular, molecular, and translational studies, this review delineates the relative significance of individual mechanisms, establishes a coherent framework for understanding glial contributions to CNS inflammation, and highlights their potential as targets for novel interventions aimed at attenuating neuroinflammation and improving clinical outcomes.

2. Glial Cell Types and Their Functions

Maintaining neural homeostasis, promoting neuronal function, and regulating synaptic activity all depend on glial cells, a varied subset of non-neuronal cells found in the CNS. Microglial cells, astrocytes, and oligodendrocytes are examples of glial cells, and each has distinct baseline functions that are essential for maintaining the integrity of the CNS [10]. An outline of their basic roles will be given in this section.

2.1. Microglial Cells

In contrast to other glial and peripheral immune cells, microglial cells are the resident innate immune cells of the CNS and are derived from primitive myeloid progenitors that invade the brain parenchyma during embryogenesis [40,41]. Microglial cells exhibit a dynamic, ramified morphology under physiological conditions. Their fine processes continuously extend and retract to actively survey the surrounding neural microenvironment. They can identify alterations in tissue homeostasis, such as indications of cellular stress, injury, infection, or changed synaptic activity, due to their motility [42]. Microglial cells can quickly shift into an activated state in response to these cues, which is characterized by morphological changes (e.g., retraction of processes and enlargement of the cell body), upregulation of surface antigens such as MHC-I (major histocompatibility complex class I) and MHC-II (major histocompatibility complex class II), and the production of reactive oxygen species (ROS), nitric oxide (NO), and some pro-inflammatory cytokines and chemokines (e.g., IL-1β, TNF-α, CCL2, and CXCL10) [43,44].
Activated microglial cells play a central role in phagocytosing apoptotic cells, cellular debris, and invading pathogens, thereby maintaining CNS integrity [45]. Crucially, they also have an impact on neurodevelopment by means of synaptic pruning, which is the process of removing weak or superfluous synaptic connections, especially during essential stages of postnatal brain maturation [46]. Through ligand-receptor pairs like CX3CL1-CX3CR1 and CD200-CD200R, microglial cells engage with neurons and other glial cells, assisting in their homeostatic, non-inflammatory state [47,48]. Many neuropathological disorders, including neurodegenerative diseases including AD, PD, and MS, are increasingly linked to dysregulation of microglial function. In these conditions, microglial cells develop persistently pro-inflammatory phenotypes that worsen neuronal damage [49].

2.2. Astrocytes

The most abundant glial cell type in the CNS, astrocytes are key specialized cells that carry out a variety of homeostatic, metabolic, and regulatory tasks necessary for healthy CNS development and operation [50]. Astrocytes exhibit a characteristic star-shaped morphology and are distinguished by the expression of GFAP. They are ideally positioned anatomically to regulate neurovascular and synaptic activity because they extend a variety of fine processes that envelop synapses, blood vessels, and other neural elements [51]. The maintenance of the BBB integrity, where perivascular astrocytic end-feet interact with pericytes and endothelial cells to control permeability and shield the CNS from circulating pathogens and toxins, is one of their most important functions [52]. In order to maintain appropriate neuronal excitability and avoid excitotoxicity, astrocytes are also essential for controlling extracellular ion concentrations, mainly K+, which they buffer via inward-rectifying K+ channels [53]. By absorbing excess neurotransmitters such as glutamate and GABA (γ-aminobutiric acid) through high-affinity transporters (such as GLT-1 -glutamate transporter 1- and GLAST -glutamate aspartate transporter-), astrocytes participate in the tripartite synapse in the synaptic domain. They then transform glutamate into glutamine, which is then recycled back to neurons [54]. Additionally, astrocytes modulate synaptic plasticity, long-term potentiation (LTP), and learning and memory processes by releasing gliotransmitters such as ATP (adenosine triphosphate), D-serine, and glutamate in a Ca2+-dependent manner [55].
Additionally, they are essential for energy metabolism because they use aerobic glycolysis to convert glucose to lactate, which is then sent to neurons via the astrocyte-neuron lactate shuttle (ANLS) [56]. Moreover, it has been reported that astroglial activation and ROS production occur during neuroinflammatory processes [57]. Astrocytes support the definition of cortical and subcortical boundaries, promote axon growth, and direct neuronal migration during development [58]. In reaction to CNS injury or disease, astrocytes undergo a process called reactive astrogliosis, which includes transcriptional and morphological alterations like hypertrophy, GFAP upregulation, and the release of inflammatory mediators [59]. Chronic astrogliosis might also prevent axonal regeneration and lead to pathological remodeling of neural circuits, even though reactive astrocytes can perform many neuroprotective tasks by creating glial scars and preventing damage from spreading [60].

2.3. Oligodendrocytes

To facilitate rapid saltatory conduction of action potentials and ensure effective neural communication over long distances, oligodendrocytes are responsible in the formation and maintenance of myelin sheaths around axons in the CNS [61]. Oligodendrocytes are derived from oligodendrocyte precursor cells (OPCs), which undergo tightly controlled processes of proliferation, migration, and differentiation to reach their mature and myelinating state. OPCs originate in the ventricular zones during embryonic development and continue into adulthood [62]. Mature oligodendrocytes produce myelin sheaths made of densely packed, multilamellar, lipid-enriched membranes by extending certain processes that enwrap discrete axonal segments. This structure promotes long-term axonal integrity and viability by facilitating saltatory propagation between nodes of Ranvier and increasing the conduction velocity of nerve impulses. It also provides trophic and metabolic support to the ensheathed axon [63].
Oligodendrocytes react to neuronal activity by adjusting the amount and thickness of myelination, which affects neural plasticity and circuit refinement. Myelination is activity-dependent [64]. In addition to their ability to myelinate, oligodendrocytes express a variety of adhesion molecules, neurotransmitter receptors (e.g., NMDA -N-methyl-D-aspartate and AMPA -α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-), and ion channels that facilitate two-way communication with astrocytes and neurons. Demyelinating disorders, like MS, are characterized by disruption of oligodendrocyte function or myelin integrity. In these disorders, immune-mediated attacks cause myelin to be lost, which results in impaired conduction, neurodegeneration, and severe neurological deficits [65]. Chronic MS and other myelin-related disorders are characterized by incomplete or dysfunctional remyelination (Figure 1), the process of regenerating new myelin sheaths from surviving OPCs [66]. Furthermore, there is growing evidence that oligodendrocyte morphology is linked to neurodevelopmental and psychiatric disorders with altered myelination patterns and white matter abnormalities, like major depressive disorder (MDD), autism spectrum disorder (ASD), and schizophrenia [67,68].

3. Mechanisms of Glial Activation in Neuroinflammation

In the CNS, glial cells are positioned as sentinels and effectors of neuroinflammatory responses through the complex and dynamic process of glial activation, which incorporates a broad range of molecular mechanisms [10,11,69]. Innate immune receptors, transcriptional reprogramming, metabolic changes, intercellular communication, and interactions with peripheral immune cells are all part of the signaling networks that control glial cells during neuroinflammation, all of which influence the progression of CNS pathologies [17,19,21].

3.1. Role of Microglial Cells in Neuroinflammation

Microglial cells (Figure 2) are uniquely adapted to sense and respond to perturbations in CNS homeostasis. Their immunosurveillance capacity relies on many germline-encoded pattern recognition receptors (PRRs), which detect some PAMPs pathogen-associated molecular pattern) and DAMPs [70,71]. TLR2 recognizes several bacterial lipoproteins and peptidoglycans [72]; TLR4 is sensitive to lipopolysaccharide (LPS) as well as endogenous ligands like heat shock proteins and high-mobility group box 1 (HMGB1) [73]; and TLR9 detects unmethylated CpG DNA (deoxyribonucleic acid) motifs [74]. TLRs (Toll-like receptors) use their Toll/interleukin-1 receptor (TIR) domains to dimerize and identify adaptor proteins after ligand engagement. With the exception of TLR3, which signals through TRIF (TIR-domain-containing adapter-inducing interferon-β), most TLRs employ MyD88 (myeloid differentiation primary response 88) as their primary adaptor. TLR4 uses both adaptors, depending on the subcellular signaling compartment [75].
MyD88 engagement promotes the recruitment of TNF receptor-associated factor 6 (TRAF6) and IL-1 receptor-associated kinases (IRAKs), promoting the activation of TAK1 (TGF-β-activated kinase 1) [76]. TAK1 subsequently phosphorylates the IκB kinase (IKK) complex (IKKα, IKKβ, and IKKγ), leading to IκBα phosphorylation and its ubiquitination and proteasomal degradation. This enables nuclear translocation of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells), typically the p65/p50 heterodimer, and transcription of several pro-inflammatory genes [77], including chemokines (CCL2, CCL5, and CXCL10), interleukins (IL-1β, IL-6, and IL-12), and TNF-α that coordinate immune cell recruitment and activation [78]. In parallel, TRIF-dependent signaling activates interferon regulatory factors (IRF3/7), inducing type I interferons (IFN-α/β) and JAK (Janus kinase)-STAT (signal transducer and activator of transcription) signaling, thus amplifying inflammation [79]. Activated microglia also upregulate MHC-I and MHC-II molecules and costimulatory proteins (CD80 and CD86), facilitating antigen-presenting cell (APC) functions, particularly under chronic neuroinflammatory conditions [80].
The NLRP3 (NOD-like receptor family pyrin domain-containing 3) inflammasome is a key downstream effector of microglial activation. Its activation requires a priming signal through TLR-NF-κB-dependent transcription of pro-IL-1β, pro-IL-18, and inflammasome components, followed by an activation signal driven by cellular stressors such as K+ efflux, ROS accumulation, mitochondrial dysfunction, or lysosomal rupture [81]. These signals induce NLRP3 oligomerization and recruitment of pro-caspase-1 and ASC (apoptosis-associated speck-like protein containing a CARD) [82]. Caspase-1 activation results in maturation and release of IL-1β and IL-18 via gasdermin D-mediated pore formation, leading to pyroptosis, a pro-inflammatory form of cell death that sustains CNS immune activation [83,84].
Extracellular ATP is a potent activator of microglial purinergic receptors, particularly the ionotropic P2X7 receptor [85]. P2X7 activation induces Ca2+ influx and K+ efflux, the latter being a critical upstream signal for NLRP3 inflammasome activation [86]. Continued P2X7 signaling promote pannexin-1-dependent pore formation, reinforcing autocrine and paracrine inflammatory loops [87]. Metabotropic purinergic receptors such as P2Y6 (UDP-responsive) and P2Y12 (ADP-responsive) regulate microglial chemotaxis and phagocytosis via PI3K/AKT/Rac1-mediated cytoskeletal remodeling, supporting targeted migration and debris clearance [88,89].
Neuron-glia crosstalk fine-tunes microglial activation. The CX3CL1-CX3CR1 axis, involving neuronal fractalkine and its microglial receptor, is essential for synaptic pruning and microglial homeostasis [90]. Disruption of this signaling mechanism promotes a pro-inflammatory microglial phenotype characterized by increased TNF-α, IL-1β, and inducible nitric oxide synthase (iNOS), thereby compromising neuroprotection [91]. Microglial activation is accompanied by metabolic reprogramming from mitochondrial oxidative phosphorylation (OXPHOS) to aerobic glycolysis [92], driven by stabilization of hypoxia-inducible factor 1-alpha (HIF-1α), which induces glycolytic enzymes and pro-inflammatory genes [93,94]. Alternatively, the activation of mTORC1 (mechanistic target of rapamycin complex 1) further supports inflammation by suppressing autophagy and enhancing biosynthetic pathways [95]. Increased flux through the pentose phosphate pathway generates NADPH, fueling NOX2 (NADPH oxidase 2)-dependent ROS production [96]. ROS regulate inflammasome activation, redox-sensitive transcription factors (NF-κB and AP-1), and oxidative damage [97]. In parallel, NF-κB- and STAT1-dependent upregulation of iNOS increases nitric oxide (NO) production, leading to peroxynitrite formation and disruption of mitochondrial, cytoskeletal, and neuronal integrity [98].
Finally, microglial expression of MHC-I, MHC-II, and costimulatory ligands such as CD40 enables interactions with CD8+ and CD4+ T cells [99]. IFN-γ released by activated T cells potently enhances microglial activation, promoting the release of neurotoxic mediators such as TNF-α, NO, and glutamate, while reinforcing MHC-II expression [100]. Sustained microglia–T cell interactions support immunological synapse formation, continuous antigen presentation, and neuroinflammation in disorders such as MS and AD [101].

3.2. Role of Astrocytes in Neuroinflammation

Regarded as support glial cells in the CNS, astrocytes are now recognized as active participants in neuroinflammation, responding to direct sensing of DAMPs and PAMPs as well as to pro-inflammatory cytokines released primarily by activated microglial cells [102,103]. Astrocytes express PRRs like TLR2, TLR3, TLR4, and RAGE (receptor for advanced glycation end products), which, upon binding ligands like HMGB1, amyloid-β, or bacterial components, trigger intracellular cascades activating transcription factors essential for astrocyte reactivity, notably the JAK-STAT and NF-κB pathways (mainly STAT3) [104,105,106].
During neuroinflammation, microglia release several cytokines (such as TNF-α and IL-1β), which activate astrocytic kinases including JAKs. Activated JAKs phosphorylate STAT3 (signal transducer and activator of transcription 3), promoting its dimerization, nuclear translocation, and binding to target gene promoters, triggering a reactive astrocyte transcriptional program that upregulates cytoskeletal proteins (e.g., GFAP, nestin, and vimentin) and induces morphological changes characteristic of astrogliosis [107,108,109]. STAT3 also enhances expression of some chemokines (CCL2, CXCL1, and CXCL10), promoting leukocyte recruitment across the BBB. Concurrently, astrocyte-derived factors regulate BBB permeability and neurovascular unit integrity, involving PKC and MAPKs to disrupt tight junction proteins (claudins and occludins), while VEGF (vascular endothelial growth factor), PGE2 (prostaglandin E2), and MMP-9 (matrix metalloproteinase 9) are upregulated, facilitating peripheral immune cell infiltration and amplifying neuroinflammatory cascades [110,111,112].
Astrocytes are crucial for glutamate homeostasis through EAAT1 (excitatory amino acid transporter 1) and EAAT2 (excitatory amino acid transporter 2) transporters, but inflammatory conditions reduce their expression and function via transcriptional repression and post-translational modifications (e.g., nitrosylation and ubiquitination), causing extracellular glutamate accumulation, excitotoxicity, Ca2+ overload, mitochondrial dysfunction, protease activation, and ROS production [113,114,115]. They also secrete damage-associated molecules such as S100B and lipocalin-2 (LCN2). S100B (S100 calcium-binding protein B) exerts trophic effects at low concentrations but acts as a DAMP at high levels via RAGE, activating NF-κB signaling. LCN2 binds to and activates 24p3R, disrupting iron homeostasis, inducing pro-inflammatory genes, and promoting cytoskeletal remodeling, reinforcing reactive glial phenotypes and neurotoxicity [116,117,118,119,120].
The phenotypic heterogeneity of astrocytes has traditionally been categorized into A1 and A2 subtypes [121]. However, Escartin et al. (2021) emphasize that astrocyte reactivity should be defined via multidimensional molecular and functional profiling, underscoring that reactive states can involve both the loss of homeostatic functions and the acquisition of novel protective or deleterious properties [122]. For the purposes of simplicity, the traditional A1/A2 classification will be used here. A1 astrocytes, induced by microglia-derived IL-1α, TNF-α, and C1q, upregulate complement components such as C3, tagging synapses for elimination by microglial CR3-mediated phagocytosis. This aberrant synaptic pruning contributes to synapse loss in neurodegenerative diseases including AD, MS, and Huntington’s disease (HD) [123,124,125,126].
Astrocyte-microglia crosstalk sustains neuroinflammation through astrocyte secretion of IL-6 and GM-CSF (granulocyte-macrophage colony-stimulating factor), which activate microglia and potentiate pro-inflammatory phenotypes, establishing a feed-forward loop [127]. This activation also upregulates adhesion molecules (ICAM-1, VCAM-1, and E-selectin) on endothelial cells, facilitating immune cell adhesion and transmigration into the CNS [128]. Astrocytes themselves can express several adhesion molecules (e.g., ICAM-1), likely modulating perivascular immune interactions, while the brain endothelium is the primary site for functional ICAM-1 upregulation [129,130].
Finally, reactive astrocytes contribute to dysregulation of CNS metabolism and redox homeostasis. In neuroinflammatory conditions, astrocytes upregulate glycolytic enzymes and attenuate mitochondrial respiration, mirroring microglial metabolic reprogramming, which leads to increased lactate production that modulates neuronal excitability and survival [131]. Astrocytes also show increased expression of NADPH oxidase isoforms, contributing to ROS production, alongside dysregulated glutathione metabolism, which compromises antioxidant defenses. This redox imbalance exacerbates cellular stress and amplifies neuroinflammation [132].

3.3. Role of Oligodendrcytes in Neuroinflammation

In contrast to microglial cells and astrocytes, which have broad classical immune functions, oligodendrocytes (primarily responsible for producing and maintaining myelin sheaths around CNS axons) [61] also actively participate in neuroinflammatory processes [22]. Their susceptibility to inflammatory mediators, oxidative stress, and metabolic disturbances contributes to oligodendrocyte dysfunction, demyelination, and propagation of inflammatory signaling in the CNS [133]. Oligodendrocyte precursor cells (OPCs), which support oligodendrocyte replacement and remyelination, express several PRRs (including TLR2 and TLR4) [134]. Binding of DAMPs or PAMPs activates intracellular cascades such as PI3K/AKT and MAPKs (ERK1/2 -extracellular signal-regulated kinases 1 and 2-, p38, and JNK -c-Jun N-terminal kinase-) [102], downregulating myelin-related genes (MBP—myelin basic protein, PLP—proteolipid protein, and MAG—myelin-associated glycoprotein) and impairing OPC maturation [135]. Transcription factors such as c-Jun and NFAT (nuclear factor of activated T-cells) further suppress differentiation, causing remyelination failure in chronic neuroinflammation [136].
Some pro-inflammatory cytokines (e.g., IFN-γ and TNF-α) activate oligodendrocytes via JAK/STAT1 and NF-κB pathways [137,138]. IFN-γ activates JAK1/JAK2 and STAT1, inducing antigen presentation genes (e.g., MHC-I) and increasing vulnerability to cytotoxic CD8+ T cells [139,140]. TNF-α activates TNFR1 (TNF receptor 1) and canonical NF-κB signaling, upregulating pro-apoptotic molecules (TRAIL—tumor necrosis factor-related apoptosis-inducing ligand and FasL—Fas ligand) and triggering caspase-mediated apoptosis [141,142].
Oligodendrocytes are susceptible to mitochondrial dysfunction during neuroinflammation due to their high metabolic demands, which are required to maintain lipid biosynthesis for myelin production. Upon activation, microglial cells and astrocytes produce and secrete reactive nitrogen species (RNS) and ROS, which induce mitochondrial membrane depolarization, promoting mitochondrial membrane permeabilization and cytochrome c release [43,143,144]. Through the formation of the apoptosome complex, which recruits and activates initiator caspase-9, this event triggers the intrinsic apoptotic pathway, which in turn triggers the activation of effector caspases-3 and -7, which cause cell death [145]. iNOS-derived NO exacerbates oligodendrocyte apoptosis and mitochondrial dysfunction [146].
On the other hand, oligodendrocytes are highly susceptible to endoplasmic reticulum (ER) stress under conditions of chronic neuroinflammation, which leads to the activation of the unfolded protein response (UPR) through the three signaling branches: PERK (protein kinase RNA-like ER kinase), IRE1α (inositol-requiring enzyme 1 alpha), and ATF6 (activating transcription factor 6) [147]. The activation of PERK induces phosphorylation of eukaryotic initiation factor 2 alpha (eIF2α), leading to an attenuation of protein synthesis while enhancing the translation of stress-responsive transcripts, such as ATF4 (activating transcription factor 4), and DDIT3 (DNA damage-inducible transcript 3), which mediate pro-apoptotic signaling pathways [148]. IRE1α mediates unconventional splicing of XBP1 (X-box binding protein 1) mRNA, enhancing expression of chaperones and ER-associated degradation (ERAD) components, but sustained IRE1α activation also induces JNK signaling, promoting oligodendrocyte apoptosis [149]. ATF6 translocates to the Golgi apparatus where it is cleaved and activated, subsequently upregulating genes involved in protein folding and quality control [150]. Persistent or unresolved ER stress disrupts myelin protein synthesis, thereby impairing myelin maintenance and repair, and may culminate in oligodendrocyte apoptosis, contributing to demyelination [151,152].
Myelin debris rich in immunogenic components, including MBP, MOG (myelin oligodendrocyte glycoprotein), and PLP, is produced by demyelination brought on by oligodendrocyte injury [152]. By stimulating microglial cells through TLR2 and TLR4 signaling and by making antigen presentation easier for peripheral immune cells, these molecules act as autoantigens, sustaining neuroinflammation [153]. The upregulation of co-stimulatory molecules (CD80/CD86) and the release of pro-inflammatory cytokines (e.g., IL-1β, TNF-α, and IL-6) are hallmarks of microglial activation in response to myelin debris, which exacerbates the local inflammatory milieu [154]. Moreover, it has been demonstrated that exosomes derived from oligodendrocytes that contain myelin antigens prime peripheral adaptive immune responses by promoting dendritic cell maturation and antigen presentation, thereby bridging the gap between systemic immunity and CNS inflammation [155].
Finally, oligodendrocytes express complement regulatory proteins like CD59; however, their expression is frequently downregulated during inflammatory stress, increasing the likelihood of complement-mediated lysis [156]. The membrane attack complex (MAC) is created when the complement cascade is activated, and it inserts into the oligodendrocyte membrane to cause cellular damage and death [157]. Complement activation products (e.g., C3a and C5a) serve as chemoattractants for infiltrating leukocytes and microglial cells, sustaining tissue damage and inflammation [158]. Another crucial factor in neuroinflammation is metabolic dysregulation in oligodendrocytes. Due to the upregulation of HIF-1α and its downstream glycolytic enzymes, such as pyruvate kinase M2 and hexokinase 2, numerous pro-inflammatory mediators cause a transition from oxidative phosphorylation to aerobic glycolysis [159]. Myelin formation and the maintenance of cellular integrity are compromised by this metabolic reprogramming, which is meant to meet energetic demands under stress but also causes lactate accumulation and reduced ATP production [160].
Oligodendrocyte-intrinsic susceptibilities, including complement-mediated cytotoxicity and metabolic reprogramming, are not isolated phenomena but are integrated within the broader glial network. Stressed or degenerating oligodendrocytes release cellular debris, pro-inflammatory cytokines, and metabolic by-products such as lactate, which function as paracrine signals modulating the activity of neighboring microglia and astrocytes. These oligodendrocyte-derived signals not only potentiate local inflammatory responses but also dynamically regulate glial activation states, establishing a self-perpetuating feed-forward loop that sustains and amplifies neuroinflammatory cascades.

3.4. Interplay Between Glial Populations

Conversely, the persistence of neuroinflammation depends on the interaction between glial populations. Cytokines, chemokines, extracellular vesicles, and metabolites like lactate and succinate all aid in microglial cells–astrocytes communication [161]. For example, succinate from activated microglial cells controls astrocyte reactivity by acting as a signaling molecule via SUCNR1 (succinate receptor 1) [162]. On the other hand, purinergic and metabotropic glutamate receptors allow astrocytic ATP and glutamate to affect microglial activation states [163,164]. In response to injury, oligodendrocytes (which are typically less interactive) secrete a variety of heat shock proteins and lipids, which are picked up by astrocytes and microglial cells and amplify the pro-inflammatory cascade [165,166]. The accumulation of oxidized lipids and misfolded proteins, such as α-synuclein, amyloid-β, and TDP-43 (TAR DNA-binding protein 43) aggregates, activates several glial pattern recognition receptors (PRRs), perpetuating chronic neuroinflammation and promoting neurodegenerative processes [167].

3.5. Additional Insights into Neuroinflammation Involving Glial Cells

The persistent activation of glial cells contributes to a maladaptive inflammatory environment that undermines neuronal integrity and function. Dysregulation of resolution pathways, including impaired production of anti-inflammatory cytokines (e.g., IL-10 and TGF-β), failure of lipid mediators such as resolvins and lipoxins, and inadequate clearance of apoptotic cells and debris through efferocytosis, prolongs glial activation and fosters a feed-forward loop of neuroinflammation [168,169]. These processes are vital to the origin and progression of some neurodegenerative and demyelinating conditions, highlighting the need for a deeper understanding of the molecular intricacies governing glial activation as a basis for developing targeted immunomodulatory therapies.
On the other hand, microglial cells and astrocytes play a key role in synaptic pruning (Figure 3), with several molecular pathways that intersect with neuroinflammatory processes [170]. Under normal developmental conditions, microglial cells mediate synaptic pruning through the complement cascade [171]. Neurons tag less active or unnecessary synapses with complement proteins (e.g., C3). Microglial cells express some complement receptors CR3 (CD11b/CD18) and CR4 (CD11c/CD18), which bind to synapses opsonized with complement fragments C3b or its inactivated form iC3b. This interaction allows the recognition, engulfment, and subsequent clearance of tagged synapses through receptor-mediated phagocytosis [172]. Astrocytes, despite their limited phagocytic capacity compared to microglial cells, contribute by secreting several cytokines (e.g., IL-33, TGF-β, and CXCL10) that modulate strongly microglial pruning activity [173]. Astrocytes also upregulate MEGF10 (MER tyrosine kinase) and MERTK (MER tyrosine kinase), two phagocytic receptors that mediate engulfment of synaptic elements under neuroinflammation [174].
In the context of neuroinflammation, microglial activation prompts the expression of complement components and phagocytic receptors [175]. This inflammatory state might lead to excessive or inappropriate synaptic pruning (Figure 3), a phenomenon implicated in disorders such as AD or MS [176,177]. Chronic inflammatory states are also associated with the depletion of several homeostatic markers (such as P2Y12 and TMEM119), favoring a pro-phagocytic phenotype that lacks the temporal and spatial precision documented during development [178]. This aberrant activation disrupts the balance of synaptic connectivity, contributing to cognitive decline and behavioral deficits associated with neuroinflammation [179].
Throughout chronic neuroinflammation, microglial activation extends beyond conventional immune functions, influencing neuronal circuitry and modulating astrocyte and oligodendrocyte activity through the secretion of several pro-inflammatory cytokines and metabolic mediators. This intercellular crosstalk establishes an intricate glial network that perpetuates neuroinflammatory responses and connects cellular dysfunction to CNS deficits, offering a conceptual framework for the study of glial interactions in neurodegenerative disorders.

4. Interplay Between Glial Cells, Neuroinflammation, and CNS Disorders

Important mediators of neuroinflammation, glial cells are increasingly recognized as context-dependent regulators of CNS homeostasis whose roles vary across disease stages and pathological environments [9]. While their early responses are often protective, persistent activation driven by disease-specific triggers and genetic susceptibility can adopt glial phenotypes toward maladaptive, self-sustaining inflammatory states, ultimately exacerbating neuronal dysfunction and degeneration [180].
In MS, glial responses are tightly coupled to autoimmune initiation and relapse-remission dynamics, with microglial cells and astrocytes acting as key amplifiers of peripheral immune infiltration rather than primary initiators of pathology. TLRs and cytokine receptors (TNFR and IFNGR) promote early microglial activation, leading to ROS, RNS, and NO production via NADPH oxidase and subsequent oxidative injury to myelin and axons [181]. As a disease progresses, sustained NLRP3 inflammasome activation reinforces chronic lesion activity, driving the release of IL-1β, IL-6, and TNF-α [182]. In parallel, microglia enhance antigen presentation via upregulation of MHC-II and CD86, thus linking innate immune activation to adaptive autoimmune responses [183]. Astrocytes can contribute in a disease-stage-dependent manner by chemokines (CCL2 and CXCL10), VEGF, and MMP-2/9, which facilitate leukocyte recruitment and blood–brain barrier disruption during active lesions [184]. Astrocytic glutamate dysregulation becomes more prominent during chronic stages, worsening excitotoxicity [185]. Oligodendrocyte apoptosis, driven by oxidative stress and immune-mediated caspase-3 activation, leads to progressive myelin protein loss and limits remyelination capacity, a hallmark of progressive MS [186].
In AD, glial activation evolves from an early protective clearance response toward a persistent maladaptive phenotype driven by aging-related factors, APOE genotype, and metabolic stress. Microglia recognize amyloid-β through TLR2/4, CD36, and TREM2, triggering prolonged NLRP3 inflammasome activation and sustained IL-1β and IL-18 release [187,188]. TREM2-dependent signaling initially supports plaque containment, but genetic variants impair this transition and favor neurotoxic inflammation. Persistent NOX2-mediated ROS and NO production further potentiate oxidative stress and compromises amyloid clearance [188]. Astrocytes undergo NF-κB-dependent reactive astrogliosis, releasing IL-6, TNF-α, and S100B, which contributes to synaptic dysfunction and neuronal apoptosis rather than acute cell loss [189]. Metabolic uncoupling becomes increasingly relevant at later stages, as reduced lactate delivery and impaired glutamate uptake undermine synaptic resilience and accelerate cognitive decline [190,191].
Neuroinflammation in ALS is closely linked to non-cell-autonomous mechanisms, in which glial dysfunction critically shapes motor neuron vulnerability. Microglia adopt a persistent pro-inflammatory M1-like phenotype, with NF-κB and MAPK activation driving secretion of IL-1β, TNF-α, NO, and prostaglandins [192,193]. This inflammatory profile intensifies with disease progression and is modulated by genetic factors such as SOD1 and C9orf72 mutations. Astrocytes progressively lose neuroprotective functions, leading to EAAT2 downregulation and glutamate-driven excitotoxicity. Moreover, astrocytes secrete neurotoxic mediators including prostaglandin E2, ROS, and TGF-β, contributing to selective motor neuron degeneration [194]. Oligodendrocytes further exacerbate pathology by reducing MCT1 expression, limiting lactate supply and compromising long-range axonal integrity during later disease stages [195].
Finally, in PD, glia-mediated oxidative and inflammatory cascades are critically influenced by environmental toxic exposures, intrinsic mitochondrial susceptibility, and α-synuclein-driven pathology. Misfolded α-synuclein activates microglial cells via TLR2/4 and NLRP3, inducing sustained ROS, TNF-α, and IL-1β production [196]. Dopaminergic neurons in the substantia nigra pars compacta are susceptible due to high basal oxidative load and NOX2-dependent microglial bursts, compounded by mitochondrial dysfunction [197]. Astrocytic dysfunction further exacerbates degeneration through reduced EAAT2-mediated glutamate uptake and impaired glutathione synthesis, compromising antioxidant defenses [198]. Loss of astrocyte-derived trophic support (influenced by GDNF and BDNF) precedes overt neuronal loss, while reactive astrocytes produce and secrete MMP-9, IL-6, and CXCL1, sustaining chronic inflammation and synaptic instability [199].
Overall, glial cells orchestrate neuroinflammatory and neurodegenerative processes in a disease-specific and temporally dynamic manner, via glial crosstalk shaping neuronal vulnerability and progression, thus highlighting glial phenotypic modulation as a promising therapeutic strategy.

5. Therapeutic Targeting of Glial Cells

Targeting glial cells has emerged as a compelling and multifaceted therapeutic strategy in the context of CNS disorders [38]. Previously viewed as ancillary to neuronal function, glial cells are now recognized as critical regulators of neuroinflammatory processes, synaptic function, myelination, and cerebral metabolism. These functional domains represent key nodes of vulnerability and opportunity across a wide spectrum of neurological diseases, such as neurodegenerative, neurodevelopmental, and neuroinflammatory conditions.
Their involvement in both the initiation and resolution of inflammation, in modulating synaptic connectivity and plasticity, and in maintaining metabolic and redox homeostasis, positions glial cells as attractive and versatile therapeutic targets. Importantly, glial dysfunction is increasingly implicated as a primary driver, rather than a secondary consequence, of CNS pathology. This has prompted a reorientation of drug development efforts toward cell-type-specific interventions aimed at modulating glial activity and phenotype.
Early trials aimed at modulating glial activity have yielded promising results, showing favorable effects on inflammatory biomarkers and, in some phase I–II studies, modest indications of clinical efficacy. However, larger-scale trials have often produced negative or inconsistent outcomes, and important safety concerns remain. Importantly, central inflammatory responses and imaging-detected side events (e.g., edema or microhemorrhages), reminiscent of amyloid-related imaging abnormalities (ARIA) reported with anti-amyloid antibodies, underscore the potential risks associated with broad modulation of immune functions in the CNS [200]. Additionally, substantial translational gaps remain, as preclinical models fail to recapitulate the cellular heterogeneity and complexity of the human glial network and do not account for the influence of aging and comorbidities [201]. Therapeutic windows and optimal dosing might differ across species, whereas existing several biomarkers often limited reliability in forecasting long-term clinical outcomes [202]. These limitations underscore the need for caution in translating glia-targeted strategies to the clinic, reinforcing the need of controlled, biomarker-guided experimental designs, cohort selection, rigorous imaging and biochemical monitoring, and stepwise dose escalation before broader implementation is considered [203]. Such approaches will help to mitigate risks and bridge the gap between preclinical insights and reproducible clinical benefit.
This section critically examines the evolving landscape of therapeutic approaches directed at glial cell populations. An overview is provided of current anti-inflammatory strategies targeting glial-mediated neuroinflammation, highlight the identification of glia-specific molecular targets and signaling pathways, and assess the potential of emerging modalities such as cell-based therapies, gene editing technologies, and glial reprogramming. On the other hand, the application of nanomedicine for the targeted delivery of pharmacological agents across the BBB is examined, with particular emphasis on those technologies that strengthen cellular specificity, minimize systemic adverse effects, and enable precise engagement of glial cells. Collectively, these approaches reflect a growing recognition of glial cells as central players in CNS homeostasis and dysfunction, and as key leverage points for next-generation therapeutic interventions.

5.1. Current Anti-Inflammatory Strategies

The majority of CNS disorders are characterized by the inflammatory response that is coordinated by glial cells, especially microglial cells and astrocytes. Chronic glial activation causes the sustained release of several pro-inflammatory cytokines (such as TNF-α, IL-1β, and IL-6), ROS, and excitotoxic agents like glutamate, which contribute to neuronal injury, even though acute inflammation can be protective.

5.1.1. Non-Steroidal Anti-Inflammatory Drugs (NSAIDs)

The potential of non-steroidal anti-inflammatory drugs (NSAIDs; Table 1), such as ibuprofen and indomethacin, to modify glial-mediated inflammation has been thoroughly studied, especially in relation to neurodegenerative diseases like AD, MS, and ALS [204,205,206,207,208,209,210,211,212]. These drugs mainly work by blocking the cyclooxygenase (COX) enzymes, namely COX-1 and COX-2, which catalyze the production of prostaglandins from arachidonic acid [213]. In the CNS, COX-2 is primarily expressed in neurons and, in pathological situations, in activated microglial cells and astrocytes [214]. Increased synthesis of pro-inflammatory prostaglandins like PGE2, which amplify glial activation, enhance cytokine production (e.g., IL-1β and TNF-α), and disrupt neuronal function and survival, is caused by the upregulation of COX-2 in neuroinflammatory states [215]. NSAIDs decrease prostaglandin levels by blocking COX-2, which can lessen excitotoxicity, oxidative stress, and glial activation [216].
Furthermore, it has been demonstrated that several NSAIDs have COX-independent effects, which could be a factor in their neuroprotective qualities [217]. One nuclear receptor implicated in anti-inflammatory signaling, peroxisome proliferator-activated receptor gamma (PPARγ), has been shown to be activated by certain NSAIDs [218]. A key pathway in the transcription of pro-inflammatory genes in microglial cells and astrocytes, NF-κB signaling, is downregulated when PPARγ is activated [219].
The clinical application of NSAIDs for neurodegenerative diseases has been mainly disappointing, despite these encouraging mechanisms seen in certain preclinical models. Their poor permeability across the BBB is a significant drawback, leading to subtherapeutic levels in the CNS [220]. The therapeutic results of NSAIDs are further complicated by their lack of cell-specificity and potential to influence both beneficial and detrimental glial responses.
Celecoxib and other COX-2-selective inhibitors were created to more accurately target inflammation and reduce the gastric side effects linked to non-selective NSAIDs [221]. However, more research is needed to validate these findings, even though their effectiveness in some clinical trials for AD has shown promise [222,223].
Table 1. Table illustrating various anti-inflammatory therapies that inhibit glial cell overactivation, including their drug family, specific compounds, molecular mechanisms of action, and associated side effects. Abbreviations: NSAID (nonsteroidal anti-inflammatory drug), COX-1 (cyclooxygenase 1), COX-2 (cyclooxygenase 2), PML (progressive multifocal leukoencephalopathy), VCAM-1 (vascular cell adhesion molecule 1), TNF-α (tumor necrosis factor-alpha), IL-1β (interleukin 1 beta), IL-6R (interleukin 6 receptor), Aβ (amyloid beta), and ARIA (amyloid-related imaging abnormalities).
Table 1. Table illustrating various anti-inflammatory therapies that inhibit glial cell overactivation, including their drug family, specific compounds, molecular mechanisms of action, and associated side effects. Abbreviations: NSAID (nonsteroidal anti-inflammatory drug), COX-1 (cyclooxygenase 1), COX-2 (cyclooxygenase 2), PML (progressive multifocal leukoencephalopathy), VCAM-1 (vascular cell adhesion molecule 1), TNF-α (tumor necrosis factor-alpha), IL-1β (interleukin 1 beta), IL-6R (interleukin 6 receptor), Aβ (amyloid beta), and ARIA (amyloid-related imaging abnormalities).
Drug
Family
CompoundMolecular
Mechanisms
Key StrengthsSide EffectsReferences
NSAIDsIbuprofen
Indomethacin
Celecoxib
Inhibition of COX-1 and COX-2
Activation of PPARγ
Widely available
Rapid anti-inflammatory
effect
Gastrointestinal bleeding
Renal dysfunction
Cardiovascular risk
Hypersensitivity
reactions
Headache
Dizziness
Renal toxicity
[213,214,215,216,218,219,224]
MinocyclineReduction in cytokine production
Modulation of apoptosis
BBB penetration
Neuroprotective potential
Dizziness
Skin pigmentation
Autoimmune hepatitis
Hypersensitivity
syndrome
[225,226,227,228,229,230]
mAbsNatalizumabBlocks α4 integrin-VCAM-1 interactionsHigh efficacy in
immune-mediated CNS diseases
PML
Hypersensitivity
reactions
Risk of infections
[231,232,233]
Infliximab
Adalimumab
Neutralization of TNF-αStrong systemic
anti-inflammatory effects
Risk of infections
Malignancies
Autoimmune responses
[234,235,236,237,238,239,240]
CanakinumabBlockade of IL-1βTargeted cytokine suppressionRisk of infections
Neutropenia
Gastrointestinal
symptoms
[241,242,243,244]
TocilizumabAntagonizes IL-6RSerum transaminase
elevation
Risk of infections
Gastrointestinal bleeding
Neutropenia
[245,246]
AducanumabInhibits Aβ plaques formationFirst approved disease-
modifying AD therapy
ARIA
Headache
Confusion
Falls
[247,248,249,250]

5.1.2. Minocycline

Using molecular mechanisms that alter cellular signaling pathways, the second-generation tetracycline antibiotic minocycline (Table 1) exhibits a pharmacological profile that goes well beyond its traditional antimicrobial activity. It also demonstrates notable anti-inflammatory and anti-apoptotic effects [225]. Minocycline mainly inhibits microglial activation, a major cause of neuroinflammation in many CNS pathologies, to produce neuroprotective effects [226]. The suppression of NF-κB and MAPK pathways causes this inhibition, which in turn reduces transcription and the release of many pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-6 [227,228].
Moreover, minocycline reduces programmed cell death in susceptible neurons by inhibiting mitochondrial cytochrome c release and subsequent caspase-3 activation, which modulates apoptotic cascades [229]. These molecular actions result in reduced lesion size, reduced BBB disruption, and preserved neuronal architecture in preclinical models of ischemic stroke, spinal cord injury, and autoimmune neuroinflammatory diseases such as MS. These outcomes all contribute to improved behavioral and functional recovery [251,252,253,254]. Long-term use of minocycline has been associated with potential negative off-target effects, including mitochondrial dysfunction, pigmentary abnormalities, and the development of autoimmune responses, despite the drug’s significant therapeutic benefits [230]. Their interactions with different cellular components outside of the CNS are probably the cause of these side effects, which emphasizes the necessity of carefully evaluating dosage regimens and long-term safety profiles in clinical applications [255].

5.1.3. Therapeutic Modulation of Cytokines: Experimental Methodologies

Monoclonal antibody (mAb) therapies have emerged as a transformative strategy in treating neuroinflammatory disorders due to their specificity in targeting pathological molecular pathways within the CNS (Table 1) [256]. A prominent example is natalizumab, an anti-α4 integrin antibody that inhibits leukocyte adhesion to VCAM-1, thus preventing immune cell migration across the BBB, a crucial mechanism in MS pathogenesis [231]. By preventing peripheral immune cell infiltration into the CNS, natalizumab modulates the neuroinflammatory milieu, resulting in decreased activation of microglial cells and astrocytes and attenuation of associated pro-inflammatory signaling pathways [232].
On the other hand, mAbs targeting TNF-α, such as infliximab and adalimumab, neutralize both soluble and membrane-bound forms of TNF-α, preventing downstream pro-inflammatory signaling [234,235]. By blocking TNF-α activity, these antibodies contribute to the complete suppression of glial activation and help mitigate the chronic neuroinflammatory responses associated with several neurodegenerative and autoimmune conditions [234,236,257]. However, these drugs compromise immunosurveillance, thereby increasing susceptibility to opportunistic infections [237,238,239,240]. Other cytokine-targeting mAbs include canakinumab, which blocks IL-1β, reducing the production of downstream mediators like IL-6 and MMPs [241,242,243,244], and tocilizumab, which antagonizes IL-6 receptor (IL-6R) signaling to dampen CNS inflammation [245,246].
In AD, therapies such as aducanumab target amyloid-beta (Aβ) plaques, promoting their clearance via microglial phagocytosis and reducing associated neuroinflammation and synaptic damage [247,248,249,250]. Finally, combination therapies targeting multiple inflammatory mediators are under evaluation to improve outcomes in some neuroinflammatory diseases (e.g., the combination of natalizumab—anti-α4 integrin, alemtuzumab—anti-CD52, ocrelizumab—anti-CD20, and daclizumab—anti-CD25 in MS [258]).

5.2. Glia-Specific Pharmacological Targets

Emerging insights into glial biology have identified some molecular targets uniquely or preferentially expressed by glial cells, opening the door to glia-specific pharmacological interventions, which hold the potential to modulate neuroinflammatory and neurodegenerative processes with greater precision while minimizing off-target effects in neurons and other CNS cell types, although their translational efficacy remains to be validated.

5.2.1. Microglial Targets

Colony-Stimulating Factor 1 Receptor (CSF1R). CSF1R is a class III receptor tyrosine kinase expressed on microglial cells and other phagocytes [259]. Activation by CSF1 or IL-34 induces receptor dimerization and autophosphorylation, triggering intracellular signaling cascades such as PI3K-AKT, MAPK/ERK, and JAK/STAT, which control microglial survival, proliferation, and differentiation [260]. Several small-molecule inhibitors (e.g., PLX3397 and GW2580) block the ATP-binding domain of CSF1R, leading to microglial depletion in numerous preclinical models [261,262]. These agents ameliorate neuroinflammation and neurodegeneration in preclinical models of AD and PD by suppressing microglial activation [263,264,265]. However, uninterrupted depletion might impair key microglial functions, like synaptic pruning, debris clearance, and neurogenesis, highlighting the therapeutic relevance of selective modulation over complete ablation (Table 2) [266].
Triggering Receptor Expressed on Myeloid Cells 2 (TREM2). TREM2 is a microglial surface receptor that signals via the adaptor DAP12 (DNAX activating protein of 12 kDa), containing ITAM (immunoreceptor tyrosine-based activation motif) motifs [267]. Ligand binding (e.g., lipids and ApoE -Apolipoprotein E-) induces DAP12 phosphorylation and activation of Syk (spleen tyrosine kinase), initiating downstream signaling via PI3K-AKT, PLCγ (phospholipase C gamma), and mTOR (mechanistic target of rapamycin) pathways [268]. TREM2 enhances phagocytosis, lipid metabolism, and cell survival, and facilitates the transition to a DAM phenotype [269]. Agonistic mAbs (e.g., AL002) that potentiate TREM2 signaling are in clinical development, aiming to promote amyloid-β clearance and shift microglial cells toward a protective, anti-inflammatory state (Table 2) [270].
Table 2. Summary of therapeutic strategies targeting glial cell hyperactivity: glial and molecular targets, pharmacological compounds, biological effects, and implications in some CNS pathologies. Abbreviations: CSF1R (colony-stimulating factor 1 receptor), ATP (adenosine triphosphate), TREM2 (triggering receptor expressed on myeloid cells 2), AD (Alzheimer’s disease), PD (Parkinson’s disease), AQP4 (aquaporin 4), NMOSD (neuromyelitis optica spectrum disorder), MCAO (middle cerebral artery occlusion), CCI (chronic constriction injury), CIPN (chemotherapy-induced peripheral neuropathy), Cx43 (connexin 43), EAAT2/GLT-1 (excitatory amino acid transporter 2/glutamate transporter 1), CNS (central nervous system), EAE (experimental autoimmune encephalomyelitis), MAI (myelin-associated inhibitor), LINGO-1 (leucine-rich repeat and Ig domain-containing Nogo receptor-interacting protein 1), and Nogo-A (neurite outgrowth inhibitor A).
Table 2. Summary of therapeutic strategies targeting glial cell hyperactivity: glial and molecular targets, pharmacological compounds, biological effects, and implications in some CNS pathologies. Abbreviations: CSF1R (colony-stimulating factor 1 receptor), ATP (adenosine triphosphate), TREM2 (triggering receptor expressed on myeloid cells 2), AD (Alzheimer’s disease), PD (Parkinson’s disease), AQP4 (aquaporin 4), NMOSD (neuromyelitis optica spectrum disorder), MCAO (middle cerebral artery occlusion), CCI (chronic constriction injury), CIPN (chemotherapy-induced peripheral neuropathy), Cx43 (connexin 43), EAAT2/GLT-1 (excitatory amino acid transporter 2/glutamate transporter 1), CNS (central nervous system), EAE (experimental autoimmune encephalomyelitis), MAI (myelin-associated inhibitor), LINGO-1 (leucine-rich repeat and Ig domain-containing Nogo receptor-interacting protein 1), and Nogo-A (neurite outgrowth inhibitor A).
Glial
Target
Molecular
Target
CompoundsBiological
Effects
CNS
Pathology
References
Microglial
cells
CSF1RPLX3397
GW2580
Reduction in neuroinflammation and neurodegeneration by inhibiting the ATP-binding site of CSF1R, which suppresses chronic microglial activation and induces microglial depletionPreclinical studies: Mouse AD (in vivo experiments)[263]
Preclinical studies: Rat PD (in vivo experiments)[264]
Preclinical studies: Mouse AD (in vivo experiments)[265]
TREM2AL002AL002, currently in clinical development, aims to enhance amyloid-β clearance and promote a protective microglial phenotypeClinical studies: Clinical trial in AD patients[266]
AstrocytesAQP4Anti-AQP4 mAbReduction in
neuroinflammation
and neurodegeneration
Preclinical studies: NMOSD rodents (in vitro and in vivo experiments)[271]
Arbidol
Tamarixetin
Preclinical studies: NMOSD mouse (in vitro and in vivo experiments)[272]
Cx43Gap19Preclinical studies: MCAO mouse (in vitro and in vivo experiments)[273]
Peptide5Preclinical studies: CCI and CIPN mouse (in vitro and in vivo experiments)[274]
RNAiPreclinical studies: Various in vitro and in vivo experiments in injured rodents[275]
EAAT2/GLT-1CeftriaxoneEAAT2/GLT-1 upregulation restores CNS homeostasis and mitigates neuroinflammation
and neurodegeneration
Preclinical studies: EAE mouse (in vitro and in vivo experiments)[276]
LDN/OSU-0212320Preclinical studies: Various in vitro and in vivo experiments in injured rodents[277]
RiluzolePreclinical studies: EAE mouse (in vitro and in vivo experiments)[278]
OligodendrocytesMAIAnti-LINGO-1
Anti-Nogo-A
Support the maturation of oligodendrocytes and repair of several demyelinated areasPreclinical studies: Various in vitro and in vivo experiments in injured rodents[279]

5.2.2. Astrocytic Targets

Aquaporin-4 (AQP4). It is the principal water channel in the CNS, localized predominantly at astrocytic end-feet through interactions with the dystrophin-associated protein complex (DAPC), including α-syntrophin, dystrophin (Dp71), and agrin [280]. This polarization is vital for regulating water flux across the BBB [281]. AQP4 mediates bidirectional water transport in response to osmotic gradients and is regulated transcriptionally and post-translationally (e.g., phosphorylation at Ser111, Thr157, and Ser180; ubiquitination) [282,283]. In CNS injury (e.g., stroke and trauma), AQP4 allows cytotoxic edema through astrocytic water influx and later contributes to vasogenic edema clearance [284]. Furthermore, AQP4 modulates neuroinflammation through cytokine signaling (such as IL-1β and TNF-α via NF-κB) and is itself regulated by STAT3-dependent signaling during reactive astrogliosis [285]. In neuromyelitis optica spectrum disorder (NMOSD), AQP4 is the main target of pathogenic IgG1 antibodies, inducing complement- and antibody-dependent cytotoxicity [286]. Therapeutic strategies (Table 2) include anti-AQP4 mAbs and some small-molecule inhibitors (e.g., arbidol and tamarixetin) [271,272].
Connexin43 (Cx43), encoded by GJA1 gene (gap junction alpha 1), forms astrocytic gap junctions via hexameric connexons, supporting intercellular exchange of ions and small metabolites for homeostatic functions [273]. While gap junctions remain open, hemichannels are typically closed but become pathologically activated by injury, ischemia, or inflammation, leading to ATP and glutamate release and promoting inflammasome activation (e.g., via P2X7 receptors) [287]. Cx43 function is modulated by phosphorylation (PKC, ERK1/2, CK1—casein kinase 1, and Src—proto-oncogene tyrosine-protein kinase Src), influencing assembly and gating [288]. Hemichannel blockade (using agents like Gap19) reduces neuroinflammation and secondary damage in models of stroke and epilepsy [289]. Other tactics include mimetic peptides (e.g., Peptide5) and RNAi (RNA interference; Table 2) [274,275].
Glutamate Transporters (EAAT2/GLT-1) are high-affinity, Na+-dependent transporters localized on perisynaptic astrocyte membranes, responsible for >90% of extracellular glutamate clearance [290]. EAAT2 activity is electrogenic, coupling glutamate uptake with Na+/H+ influx and K+ efflux [291]. Its expression is highly regulated via NF-κB and CREB (cAMP response element-binding) pathways [292]. Under inflammation, EAAT2 is downregulated through epigenetic and proteasomal mechanisms, contributing to excitotoxicity [293]. EAAT2 dysfunction is strongly associated with ALS, AD, HD, and epilepsy [294,295,296,297]. Therapeutic upregulation strategies include β-lactams (e.g., ceftriaxone), small-molecule enhancers (e.g., LDN/OSU-0212320), and riluzole (Table 2) [276,277,278].

5.2.3. Oligodendroglial Targets

Myelin-associated inhibitors (MAIs), like Nogo-A and LINGO-1, negatively regulate remyelination in the CNS by activating the Nogo receptor 1 (NgR1) with co-receptors such as p75NTR, TROY (tumor necrosis factor receptor superfamily member 19), and LINGO-1 [298]. This activation triggers the RhoA (Ras homolog family member A)/ROCK (Rho-associated coiled-coil-containing protein kinase) signaling pathway, which inhibits cytoskeletal remodeling necessary for axonal growth and OPC differentiation [279]. LINGO-1 acts as a crucial co-receptor mediating suppression of OPC maturation [299]. Therapeutic agents targeting Nogo-A or LINGO-1 aim to promote oligodendrocyte differentiation and myelin repair, particularly in MS (Table 2) [279].
Monocarboxylate transporter 1 (MCT1), mainly expressed on oligodendrocytes, mediates the transport of lactate and pyruvate, encouraging metabolic coupling between oligodendrocytes and neurons [300]. Oligodendrocytes convert glucose to lactate, which is shuttled through MCT1 to axons, where neurons uptake it through MCT2 (monocarboxylate transporter 2) to support mitochondrial energy production [301]. Impaired MCT1 function in those demyelinating diseases disrupts metabolic coupling between oligodendrocytes and neurons, leading to energy deficits and degeneration [302]. Improving MCT1 activity constitutes a potential therapeutic strategy to restore neuronal metabolic support and prevent axonal loss in MS [303].

5.3. Gene Therapies

Innovative strategies involving gene editing technologies are emerging as powerful tools with unprecedented potential to reprogram dysfunctional glial populations. These approaches not only enable targeted manipulation of glial cell identity and function, but also open new avenues for treating a wide range of neurodegenerative and neurodevelopmental disorders where glial dysfunction plays a key role [304].

5.3.1. Viral Vector-Based Gene Delivery

Adeno-associated viruses (AAVs) have emerged as the vectors of choice for in vivo gene delivery due to their low immunogenicity, long-term expression, and capacity for tissue-specific targeting (Table 3) [305]. Recent findings in AAV engineering have enabled the use of glial-specific promoters (e.g., GFAP, Iba1—ionized calcium binding adapter molecule 1, and MBP) to drive selective expression of therapeutic genes in specific glial subpopulations [306,307]. This precision targeting is valuable for modulating the glial contribution to neurodegenerative and neuroinflammatory pathologies. For instance, AAV-mediated overexpression of neuroprotective molecules including IGF-1 (insulin-like growth factor 1) or BDNF in astrocytes have been shown to confer neuroprotection, reduce excitotoxicity, and support synaptic integrity [308,309]. Concurrently, the delivery of genetic material to microglial cells through tailored AAV vectors shift these cells toward a homeostatic, anti-inflammatory phenotype, thus mitigating chronic neuroinflammation and associated neurodegeneration [310]. Current studies aim to optimize vector serotypes and promoter designs aimed at improving gene transfer efficiency, target-cell specificity, and biosafety, with the aim of enhancing their applicability to translational medicine [311].

5.3.2. RNA-Based Therapies

Small interfering RNAs (siRNAs) and antisense oligonucleotides (ASOs) are two examples of RNA (ribonucleic acid)-targeting modalities that provide tools for post-transcriptional modification of gene expression in glial cells (Table 3). These molecules can be designed to suppress the translation of deleterious proteins, restore normal splicing patterns, or selectively target and degrade mutant transcripts [312,313]. ASOs (targeting Sod1 -superoxide dismutase 1- and/or C9orf72) transcripts have been developed in the context of ALS to lessen the buildup of toxic proteins and RNA foci in neurons, astrocytes, and microglial cells, which contribute to the progression of the disease [314,315]. ASOs are especially well-suited for repeated therapeutic administration due to their stability and capacity to cross the BBB [316]. Moreover, glial-derived inflammatory mediators and oxidative stress-related enzymes have been demonstrated to be downregulated by siRNAs [317]. With many RNA-based treatments currently making their way via clinical trials for different CNS disorders, these therapeutic options represent a rapidly developing frontier in glia-targeted therapy.

5.4. Cell Replacement Therapy

5.4.1. Astrocyte Transplantation

The transplantation of astrocyte precursors has emerged as a promising therapeutic approach in models of neurodegenerative and traumatic CNS disorders, mainly SCI and ALS (Table 4) [318,319]. Preclinical studies have shown that the introduction of astrocyte precursor cells into the injured spinal cord improves neuronal viability, reduces gliosis, and allows axonal regeneration [320,321]. An important advancement in this field is the derivation of astrocytes from human induced pluripotent stem cells (iPSCs), which are currently under investigation for their safety and efficacy in several clinical settings [322]. These iPSC-derived astrocytes not only exhibit functional properties consistent with endogenous astrocytes but also hold the potential for personalized, autologous cell therapy strategies [323].

5.4.2. OPC Transplantation

OPC transplantation represents a novel strategy to restore myelination in several demyelinating diseases including MS and in the context of traumatic SCI (Table 4) [324]. In preclinical models, transplanted OPCs have been shown to migrate to those demyelinated regions, differentiate into mature oligodendrocytes, and effectively remyelinate axons [325,326]. This remyelination contributes to the restoration of nerve conduction velocities and amelioration of functional deficits [327]. iPSC-derived OPCs have shown promising results in terms of scalability, reproducibility, and potential for clinical translation [328].

5.4.3. Microglial Transplantation

Microglial replacement therapy is an emerging experimental approach intended for reestablishing a balanced neuroimmune environment within the CNS (Table 4). Dysregulation of microglial function is implicated in many neurological disorders, such as neurodegenerative diseases and CNS injuries. Some therapeutic strategies for replacing dysfunctional microglial cells include the transplantation of hematopoietic stem cells, which can differentiate into microglia-like cells within the CNS, as well as the transplantation of microglia derived from human iPSCs, offering a targeted and physiologically relevant approach to restoring microglial function [329].
Recent studies using mouse models with human CSF1R mutations demonstrated that microglial depletion followed by replacement with healthy bone marrow-derived microglial cells (Mr BMT) restored myelin integrity, reduced axonal spheroids, and improved cognitive function. This approach was applied to eight ALSP (adult-onset leukoencephalopathy with axonal spheroids and pigmented glia) patients using conventional bone marrow transplantation, resulting in stabilization of disease progression over a 24-month period [330]. Moreover, direct intracerebral injection of Sca1 (stem cell antigen-1 negative) myeloid progenitor cells has been shown to efficiently repopulate the microglial compartment in the absence of myeloablative preconditioning, thus providing proof-of-concept for microglial replacement. This approach rescued a mouse model of Sandhoff disease and highlighted the potential of human iPSC-derived progenitors as an alternative to conventional hematopoietic stem cell transplantation [331]. The replacement of dysfunctional microglial cells with peripherally derived microglia-like cells (MLCs) renews CNS homeostasis, constituting an alternative therapeutic strategy for neurodegenerative pathologies associated with GM2 (GM2 ganglioside) accumulation, including Sandhoff disease [332].
While these interventions are still in early stages of development, preliminary studies suggest that appropriately engrafted microglial cells can integrate into the CNS tissues, assume homeostatic phenotypes, and modulate several inflammatory responses [333]. Further investigations are required to optimize delivery methods, ensure long-term engraftment, and assess therapeutic efficacy in clinical settings [334].

5.5. Nanomedicine and Targeted Drug Delivery

The BBB continues to represent a major impediment to the effective delivery of therapeutic agents to CNS [335]. Its highly selective permeability restricts the passage of most pharmacological compounds, thereby limiting treatment efficacy for numerous neurological disorders. In this context, the field of nanotechnology offers a transformative strategy to overcome the BBB’s restrictive properties [336]. Nanocarriers can be engineered to traverse the barrier via receptor-mediated transport or other mechanisms, enabling precise delivery of therapeutic payloads [337]. Additionally, these nanosystems (Table 5) offer the potential for cell-type-specific targeting, like selective delivery to glial cells, thus enhancing treatment specificity while minimizing off-target effects [338].
Polymeric nanoparticles (PNPs) and liposomes are effective nanocarriers that can encapsulate a variety of therapeutic agents, such as peptides, nucleic acids (such as plasmid DNA, siRNA, and miRNA—microRNA), and anti-inflammatory medications [339]. Targeting ligands like transferrin or the rabies virus glycoprotein (RVG) peptide can be added to the surface of these nanocarriers to functionalize them. These ligands attach to the receptors that are widely expressed on the BBB’s endothelial cells, like the transferrin receptor or the nicotinic acetylcholine receptor [340,341]. This makes it easier for the nanoparticles to cross the blood–brain barrier through receptor-mediated transcytosis. Through ligand-receptor interactions with glial cell markers (e.g., CD44 for astrocytes and CD11b for microglial cells), other cell-specific targeting can be accomplished after infiltrating the CNS parenchyma [342,343]. Therapeutic agents are released into the intracellular compartment upon cellular uptake, usually by clathrin or caveolin-mediated endocytosis. By disrupting pathways like NF-κB, JAK/STAT, or MAPK, these agents can exert gene-modulatory or anti-inflammatory effects [344,345].
Extracellular vesicles (EVs), such as exosomes, especially those originating from mesenchymal stem cells (MSCs) or engineered cell lines, have the innate ability to cross the BBB through endogenous pathways like adsorptive-mediated transcytosis or integrin-mediated adhesion and uptake [346]. Glial cells can more effectively recognize and internalize them due to their membrane biochemical composition, which is enriched in tetraspanins (such as CD63 and CD81) and integrins [347]. Through electroporation or endogenous sorting signals (e.g., EXOmotifs or exosomal motifs), engineered exosomes can be loaded with therapeutic proteins or regulatory miRNAs (like miR-124-3p) [348,349,350]. The exosomal contents, once internalized by glial cells, act on intracellular signaling cascades such as TLR4/NF-κB to influence neuroinflammatory signaling, promote neuroprotection, or rewire glial phenotypes [351]. Their immunological tolerance and physiological origin enable safe, repeated, and long-term therapeutic application in neurological disorders [352].
On the other hand, the BrainShuttleTM platform employs engineered antibodies that bind to receptors on BBB endothelial cells, such as the transferrin receptor, and are actively transported into the CNS through receptor-mediated transcytosis. Following translocation across the BBB, therapeutic agents can interact with microglial cells and regulate their inflammatory signaling pathways [353]. Moreover, nanobodies (small antibody fragments derived from camelid antibodies) are advantageous due to their high stability, small size, and ability to recognize unique epitopes [354]. Nanobodies might represent an excellent strategy to target various molecules expressed in glial cells, aiming to enhance their functionality in the context of neuroinflammation [355].
Finally, Zhu et al. showed that topoisomerase 1 (TOP1) inhibitors, such as camptothecin and topotecan, suppress microglial and macrophage inflammation in vitro and reduce neuroinflammation in vivo. Furthermore, a β-glucan-coated DNA origami nanocarrier (TopoGami) was developed to deliver topotecan selectively to myeloid cells. This intervention suppressed microglial activation and attenuated disease progression in a MS-like model, underscoring the therapeutic potential of targeted TOP1 inhibition in neuroinflammatory disorders [356].

6. Conclusions

Beyond their historically assigned supporting roles in the CNS, glial cells are recognized as important regulators of neuroinflammation. While a substantial body of experimental work has clarified many aspects of glial activation, intercellular communication, and their involvement in neuropathological conditions, the strength and generalizability of this evidence vary considerably across model systems and pathological contexts. Much of the current mechanistic understanding derives from well-controlled animal models and in vitro studies, which have been instrumental in dissecting signaling pathways but do not always recapitulate the complexity and/or cellular diversity of the human CNS. Consequently, interpretations of glial-mediated processes must be framed with appropriate caution.
Glial intercellular communication constitutes a tightly regulated system that can promote tissue repair and homeostasis or, when dysregulated, contribute to neurodegeneration. However, the causal relationships between specific signaling pathways and defined pathological outcomes are often inferred rather than directly demonstrated, and their reproducibility across laboratories and disease models remains heterogeneous. In particular, context-dependent variables like species, developmental stage, brain region, and injury paradigm substantially influence observed outcomes.
Microglial activation encompasses a spectrum of phenotypic states ranging from pro-inflammatory and potentially neurotoxic responses to homeostatic and neuroprotective surveillance functions. On the other hand, reactive astrocytes adopt heterogeneous functional states that influence synaptic remodeling, BBB integrity, and neuronal survival. Although these concepts are supported by numerous transcriptomic and functional studies, the frequently cited phenotype classifications remain operational frameworks rather than universally validated biological entities, and their relevance to human disease is still under active investigation. These findings underscore the huge importance of conceptualizing glial plasticity, rather than relying on simplified or binary activation models.
Reciprocal interactions between microglia and astrocytes play a central role in shaping the neuroinflammatory cascade. Experimental evidence indicates that such interactions can amplify inflammatory signaling and sustain neuronal damage, but also facilitate debris clearance, trophic support, and inflammation resolution under specific regulatory conditions. Notably, the balance between these opposing outcomes is highly model-dependent, and direct evidence for analogous mechanisms operating in human neurodegenerative disorders remains limited. OPCs are increasingly recognized as immune-responsive cells capable of sensing pro-inflammatory mediators and modulating local immune environments; however, this emerging role is largely supported by preclinical studies and awaits rigorous validation in vivo and in human tissue.
The growing insights into glial biology have catalyzed a paradigm shift in CNS-focused therapeutic strategies. Targeting glial cells offers several opportunities to modulate immune regulation, metabolic homeostasis, and synaptic repair. Despite these advances, the clinical translation of glia-directed therapies has been constrained by issues like limited cell-type specificity, off-target effects, and insufficient consideration of the temporal and spatial dynamics of glial activation. Emerging approaches (e.g., glia-targeted molecular therapies, genome editing technologies, cell-based interventions, and nanotechnology-enabled delivery platforms) offer promising avenues, but most remain at early or preclinical stages of development. Importantly, claims regarding therapeutic efficacy should be interpreted cautiously until supported by reproducible, longitudinal, and clinically relevant data. The identification and robust validation of biomarkers capable of discriminating distinct glial states will be essential for advancing precision medicine approaches in CNS disorders [319], yet such biomarkers are currently scarce and incompletely validated.
Despite notable progress, significant limitations constrain the current understanding of glial biology. A large proportion of available evidence is derived from animal models that only partially reproduce the molecular, cellular, and clinical features of human CNS diseases. In vitro systems, while valuable for mechanistic dissection, often employ reductionist conditions that fail to capture the full diversity and plasticity of glial responses in vivo. Furthermore, technical variability, batch effects, and analytical challenges in single-cell and spatial transcriptomics complicate cross-study comparisons and limit reproducibility. These constraints highlight the need for longitudinal, cross-species, and integrative research frameworks capable of bridging mechanistic insights with clinical relevance.
Taken together, the literature supports several overarching principles in glial-driven neuroinflammation, while also revealing important gaps in knowledge. Neuroinflammatory processes should be viewed as emergent properties of coordinated multicellular glial networks rather than isolated and cell-autonomous responses. Glial activation is intrinsically context-dependent, influenced by temporal stage, regional identity, disease etiology, and systemic factors, pointing out the limitations of classification frameworks. Moreover, bidirectional communication between glial cells and neurons, as well as among different glial subtypes, represents a critical regulatory axis that determines whether inflammatory signaling culminates in repair or degeneration. Notably, the relative contribution of each component of this axis remains incompletely resolved in human disease settings.
In summary, glial cells (Figure 4) are key regulators of both the initiation and resolution of neuroinflammation, necessitating a shift in therapeutic strategies that recognizes them as active modulators of CNS immunity and plasticity. Future research should focus on comprehensive characterization of glial states across the lifespan and disease spectrum, delineation of conserved versus disease-specific signaling networks, and integration of multi-omic, spatial, and longitudinal data. Translational, systems-level studies will be critical to differentiate robust mechanisms from context-dependent observations and to fully realize the therapeutic potential of glial cells.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The author declares no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
24p3R24p3 receptor
A1Astrocyte subtype 1
A2Astrocyte subtype 2
AAVAdeno-associated virus
ADAlzheimer’s disease
AKTProtein kinase B
ALSAmyotrophic lateral sclerosis
ALSPAdult-onset leukoencephalopathy with axonal spheroids and pigmented glia
AMPAα-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
ANLSAstrocyte-neuron lactate shuttle
AP-1Activator protein 1
APCAntigen-presenting cell
ApoEApolipoprotein E
AQP4Aquaporin 4
Arg1Arginase 1
ARIAAmyloid-related imaging abnormalities
ASCApoptosis-associated speck-like protein containing a CARD
ASDAutism spectrum disorder
ASOAntisense oligonucleotide
ATF4Activating transcription factor 4
ATF6Activating transcription factor 6
ATPAdenosine triphosphate
Amyloid beta
BBBBlood–brain barrier
BDNFBrain-derived neurotrophic factor
C1qComplement component 1q
C3Complement component 3
C3aComplement component 3a
C3bComplement component 3b
C5aComplement component 5a
C9orf72Chromosome 9 open reading frame 72
CARDCaspase activation and recruitment domain
CCIChronic constriction injury
CCL2CC chemokine ligand 2
CCL5CC chemokine ligand 5
CD11bCluster of differentiation 11b
CD11cCluster of differentiation 11c
CD16Cluster of differentiation 16
CD18Cluster of differentiation 18
CD20Cluster of differentiation 20
CD200Cluster of differentiation 200
CD200RCluster of differentiation 200 receptor
CD206Cluster of differentiation 206
CD25Cluster of differentiation 25
CD32Cluster of differentiation 32
CD36Cluster of differentiation 36
CD4Cluster of differentiation 4
CD40Cluster of differentiation 40
CD44Cluster of differentiation 44
CD52Cluster of differentiation 52
CD55Cluster of differentiation 55
CD59Cluster of differentiation 59
CD8Cluster of differentiation 8
CD80Cluster of differentiation 80
CD86Cluster of differentiation 86
CIPNChemotherapy-induced peripheral neuropathy
CK1Casein kinase 1
CNSCentral nervous system
COXCyclooxygenase
CR3Complement receptor 3
CR4Complement receptor 4
CREBcAMP response element-binding
CSF1Colony-stimulating factor 1
CSF1RColony-stimulating factor 1 receptor
CX3CL1Chemokine (C-X3-C motif) ligand 1
CX3CR1Chemokine (C-X3-C motif) receptor 1
Cx43Connexin 43
CXCL1Chemokine (C-X-C motif) ligand 1
CXCL10Chemokine (C-X-C motif) ligand 10
DAMDisease-associated microglia
DAMPDamage-associated molecular pattern
DAP12DNAX activating protein of 12 kDa
DAPCDystrophin-associated protein complex
DDIT3DNA damage-inducible transcript 3
DNADeoxyribonucleic acid
Dp71Dystrophin protein of 71 kDa
EAAT1Excitatory amino acid transporter 1
EAAT2Excitatory amino acid transporter 2
EAEExperimental autoimmune encephalomyelitis
eIF2αEukaryotic initiation factor 2 alpha
EREndoplasmic reticulum
ERADER-associated degradation
ERK1/2Extracellular signal-regulated kinases 1 and 2
EVExtracellular vesicle
EXOmotifExosomal sorting motif
FasLFas ligand
Fizz1Found in inflammatory zone 1
GABAGamma-aminobutyric acid
GDNFGlial cell line-derived neurotrophic factor
GFAPGlial fibrillary acidic protein
GJA1Gap junction alpha 1
GLASTGlutamate aspartate transporter
GLT-1Glutamate transporter 1
GM2GM2 ganglioside
GM-CSFGranulocyte-macrophage colony-stimulating factor
HDHuntington’s disease
HIF-1αHypoxia-inducible factor 1 alpha
HMGB1High-mobility group box 1
Iba1Ionized calcium binding adapter molecule 1
iC3bInactive complement component 3b
ICAM-1Intercellular adhesion molecule 1
IFN-α/βInterferon alpha/beta
IFN-γInterferon gamma
IFNGRInterferon-γ receptor
IGF-1Insulin-like growth factor 1
IgGImmunoglobulin G
IgG1Immunoglobulin G subtype 1
IKKIκB kinase
IKKαIκB kinase alpha
IKKβIκB kinase beta
IKKγIκB kinase gamma
IL-10Interleukin 10
IL-12Interleukin 12
IL-18 Interleukin 18
IL-1αInterleukin 1 alpha
IL-1βInterleukin 1 beta
IL-33Interleukin 33
IL-34Interleukin 34
IL-4Interleukin 4
IL-6Interleukin 6
IL-6RInterleukin 6 receptor
iNOSInducible nitric oxide synthase
IRAKInterleukin-1 receptor-associated kinase
IRE1αInositol-requiring enzyme 1 alpha
IRF3/7Interferon regulatory factors 3 and 7
ITAMImmunoreceptor tyrosine-based activation motif
IκBαInhibitor of κB alpha
JAKJanus kinase
JAK/STATJanus kinase/signal transducer and activator of transcription
JNKc-Jun N-terminal kinase
LCN2Lipocalin 2
LINGO-1Leucine-rich repeat and Ig domain-containing Nogo receptor-interacting protein 1
LPSLipopolysaccharide
LTPLong-term potentiation
M1Microglia subtype 1
M2Microglia subtype 2
mAbMonoclonal antibody
MACMembrane attack complex
MAGMyelin-associated glycoprotein
MAIMyelin-associated inhibitor
MAPKMitogen-activated protein kinase
MBPMyelin basic protein
MCT1Monocarboxylate transporter 1
MCT2Monocarboxylate transporter 2
MDDMajor depressive disorder
MEGF10Multiple EGF-like domains 10
MERTKMER tyrosine kinase
MHC-IMajor histocompatibility complex class I
MHC-IIMajor histocompatibility complex class II
miRNAMicroRNA
MCAOMiddle cerebral artery occlusion
MLCMicroglia-like cell
MMP-2Matrix metalloproteinase 2
MMP-9Matrix metalloproteinase 9
MOGMyelin oligodendrocyte glycoprotein
Mr BMTMicroglia replacement by bone marrow transplantation
MSMultiple sclerosis
MSCMesenchymal stem cell
mTORMechanistic target of rapamycin
mTORC1Mechanistic target of rapamycin complex 1
MyD88Myeloid differentiation primary response 88
NADPHNicotinamide adenine dinucleotide phosphate
NFATNuclear factor of activated T-cells
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
NgR1Nogo receptor 1
NLRP3NOD-like receptor family pyrin domain-containing 3
NMDAN-methyl-D-aspartate
NMOSDNeuromyelitis optica spectrum disorder
NONitric oxide
Nogo-ANeurite outgrowth inhibitor A
NOX2NADPH oxidase 2
NSAIDNon-steroidal anti-inflammatory drug
OPCOligodendrocyte precursor cell
OXPHOSOxidative phosphorylation
P2X7Purinergic receptor P2X, ligand-gated ion channel 7
P2Y12Purinergic receptor P2Y12
P2Y6Purinergic receptor P2Y6
p38p38 protein
p75NTRp75 neurotrophin receptor
PAMPPathogen-associated molecular pattern
PDParkinson’s disease
PERKProtein kinase RNA-like ER kinase
PGE2Prostaglandin E2
PI3KPhosphoinositide 3-kinase
PKCProtein kinase C
PLCγPhospholipase C gamma
PLPProteolipid protein
PLP1Proteolipid protein 1
PMLProgressive multifocal leukoencephalopathy
PNPPolymeric nanoparticle
PPARγPeroxisome proliferator-activated receptor gamma
PRRPattern recognition receptor
Rac1Ras-related C3 botulinum toxin substrate 1
RAGEReceptor for advanced glycation end products
RhoARas homolog family member A
RNARibonucleic acid
RNAiRNA interference
RNSReactive nitrogen species
ROCKRho-associated coiled-coil-containing protein kinase
ROSReactive oxygen species
RVGRabies virus glycoprotein
S100BS100 calcium-binding protein B
scRNA-seqSingle-cell RNA sequencing
Sca1Stem cell antigen-1 negative
siRNASmall interfering RNA
SIRP1ASignal regulatory protein alpha
SOCS3Suppressor of cytokine signaling 3
SOD1Superoxide dismutase 1
SrcProto-oncogene tyrosine-protein kinase Src
STATSignal transducer and activator of transcription
STAT1Signal transducer and activator of transcription 1
SUCNR1Succinate receptor 1
SykSpleen tyrosine kinase
TAK1TGF-β activated kinase 1
TBITraumatic brain injury
TDP-43TAR DNA-binding protein 43
TGF-βTransforming growth factor beta
TIRToll/interleukin-1 receptor
TLRToll-like receptor
TLR2Toll-like receptor 2
TLR3Toll-like receptor 3
TLR4Toll-like receptor 4
TLR9Toll-like receptor 9
TMEM119Transmembrane protein 119
TNFRTumor necrosis factor receptor
TNFR1Tumor necrosis factor receptor 1
TNF-αTumor necrosis factor alpha
TOP1Topoisomerase 1
TRAF6TNF receptor-associated factor 6
TRAILTumor necrosis factor-related apoptosis-inducing ligand
TREM2Triggering receptor expressed on myeloid cells 2
TRIFTIR-domain-containing adapter-inducing interferon
TROYTumor necrosis factor receptor superfamily member 19
UPRUnfolded protein response
VCAM-1Vascular cell adhesion molecule 1
VEGFVascular endothelial growth factor
XBP1X-box binding protein 1
Ym1Chitinase-like protein 3

References

  1. Kölliker-Frers, R.; Udovin, L.; Otero-Losada, M.; Kobiec, T.; Herrera, M.I.; Palacios, J.; Razzitte, G.; Capani, F. Neuroinflammation: An Integrating Overview of Reactive-Neuroimmune Cell Interactions in Health and Disease. Mediat. Inflamm. 2021, 2021, 9999146. [Google Scholar] [CrossRef] [PubMed]
  2. García-Domínguez, M. Neuroinflammation: Mechanisms, Dual Roles, and Therapeutic Strategies in Neurological Disorders. Curr. Issues Mol. Biol. 2025, 47, 417. [Google Scholar] [CrossRef] [PubMed]
  3. Xiong, Y.; Mahmood, A.; Chopp, M. Current understanding of neuroinflammation after traumatic brain injury and cell-based therapeutic opportunities. Chin. J. Traumatol. 2018, 21, 137–151. [Google Scholar] [CrossRef] [PubMed]
  4. Alsbrook, D.L.; Di Napoli, M.; Bhatia, K.; Biller, J.; Andalib, S.; Hinduja, A.; Rodrigues, R.; Rodriguez, M.; Sabbagh, S.Y.; Selim, M.; et al. Neuroinflammation in Acute Ischemic and Hemorrhagic Stroke. Curr. Neurol. Neurosci. Rep. 2023, 23, 407–431. [Google Scholar] [CrossRef]
  5. Heneka, M.T.; van der Flier, W.M.; Jessen, F.; Hoozemanns, J.; Thal, D.R.; Boche, D.; Brosseron, F.; Teunissen, C.; Zetterberg, H.; Jacobs, A.H.; et al. Neuroinflammation in Alzheimer disease. Nat. Rev. Immunol. 2025, 25, 321–352. [Google Scholar] [CrossRef]
  6. Çınar, E.; Tel, B.C.; Şahin, G. Neuroinflammation in Parkinson’s Disease and its Treatment Opportunities. Balk. Med. J. 2022, 39, 318–333. [Google Scholar] [CrossRef]
  7. Bjelobaba, I.; Savic, D.; Lavrnja, I. Multiple Sclerosis and Neuroinflammation: The Overview of Current and Prospective Therapies. Curr. Pharm. Des. 2017, 23, 693–730. [Google Scholar] [CrossRef]
  8. Calma, A.D.; Pavey, N.; Menon, P.; Vucic, S. Neuroinflammation in amyotrophic lateral sclerosis: Pathogenic insights and therapeutic implications. Curr. Opin. Neurol. 2024, 37, 585–592. [Google Scholar] [CrossRef]
  9. Afridi, R.; Tsuda, M.; Ryu, H.; Suk, K. The Function of Glial Cells in the Neuroinflammatory and Neuroimmunological Responses. Cells 2022, 11, 659. [Google Scholar] [CrossRef]
  10. Jäkel, S.; Dimou, L. Glial Cells and Their Function in the Adult Brain: A Journey through the History of Their Ablation. Front. Cell. Neurosci. 2017, 11, 24. [Google Scholar] [CrossRef]
  11. Allen, N.J.; Lyons, D.A. Glia as architects of central nervous system formation and function. Science 2018, 362, 181–185. [Google Scholar] [CrossRef] [PubMed]
  12. Afridi, R.; Kim, J.H.; Rahman, M.H.; Suk, K. Metabolic Regulation of Glial Phenotypes: Implications in Neuron-Glia Interactions and Neurological Disorders. Front. Cell. Neurosci. 2020, 14, 20. [Google Scholar] [CrossRef] [PubMed]
  13. Fields, R.D.; Woo, D.H.; Basser, P.J. Glial Regulation of the Neuronal Connectome through Local and Long-Distant Communication. Neuron 2015, 86, 374–386. [Google Scholar] [CrossRef] [PubMed]
  14. Parpura, V.; Heneka, M.T.; Montana, V.; Oliet, S.H.; Schousboe, A.; Haydon, P.G.; Stout, R.F., Jr.; Spray, D.C.; Reichenbach, A.; Pannicke, T.; et al. Glial cells in (patho)physiology. J. Neurochem. 2012, 121, 4–27. [Google Scholar] [CrossRef]
  15. Colonna, M.; Butovsky, O. Microglia Function in the Central Nervous System During Health and Neurodegeneration. Annu. Rev. Immunol. 2017, 35, 441–468. [Google Scholar] [CrossRef]
  16. Wendimu, M.Y.; Hooks, S.B. Microglia Phenotypes in Aging and Neurodegenerative Diseases. Cells 2022, 11, 2091. [Google Scholar] [CrossRef]
  17. Woodburn, S.C.; Bollinger, J.L.; Wohleb, E.S. The semantics of microglia activation: Neuroinflammation, homeostasis, and stress. J. Neuroinflamm. 2021, 18, 258. [Google Scholar] [CrossRef]
  18. Parpura, V.; Verkhratsky, A. Homeostatic function of astrocytes: Ca(2+) and Na(+) signalling. Transl. Neurosci. 2012, 3, 334–344. [Google Scholar] [CrossRef]
  19. Matusova, Z.; Hol, E.M.; Pekny, M.; Kubista, M.; Valihrach, L. Reactive astrogliosis in the era of single-cell transcriptomics. Front. Cell. Neurosci. 2023, 17, 1173200. [Google Scholar]
  20. Lee, H.G.; Lee, J.H.; Flausino, L.E.; Quintana, F.J. Neuroinflammation: An astrocyte perspective. Sci. Transl. Med. 2023, 15, eadi7828. [Google Scholar] [CrossRef]
  21. López-Muguruza, E.; Peiró-Moreno, C.; Pérez-Cerdá, F.; Matute, C.; Ruiz, A. Del Río Hortega’s insights into oligodendrocytes: Recent advances in subtype characterization and functional roles in axonal support and disease. Front. Neuroanat. 2025, 19, 1557214. [Google Scholar] [CrossRef] [PubMed]
  22. Boccazzi, M.; Raffaele, S.; Fumagalli, M. Not only myelination: The immune-inflammatory functions of oligodendrocytes. Neural Regen. Res. 2022, 17, 2661–2663. [Google Scholar] [CrossRef]
  23. Yalçın, B.; Monje, M. Microenvironmental interactions of oligodendroglial cells. Dev. Cell. 2021, 56, 1821–1832. [Google Scholar] [CrossRef] [PubMed]
  24. Bernaus, A.; Blanco, S.; Sevilla, A. Glia Crosstalk in Neuroinflammatory Diseases. Front. Cell. Neurosci. 2020, 14, 209. [Google Scholar] [CrossRef] [PubMed]
  25. Virtuoso, A.; De Luca, C.; Korai, S.A.; Papa, M.; Cirillo, G. Neuroinflammation and glial activation in the central nervous system: A metabolic perspective. Neural Regen. Res. 2023, 18, 1025–1026. [Google Scholar] [CrossRef]
  26. Quan, L.; Uyeda, A.; Muramatsu, R. Central nervous system regeneration: The roles of glial cells in the potential molecular mechanism underlying remyelination. Inflamm. Regen. 2022, 42, 7. [Google Scholar] [CrossRef]
  27. Afridi, R.; Suk, K. Targeting glial metabolism in neurodegenerative diseases: Detail matters. Neural Regen. Res. 2023, 18, 2393–2394. [Google Scholar] [CrossRef]
  28. Spurgat, M.S.; Tang, S.-J. Single-Cell RNA-Sequencing: Astrocyte and Microglial Heterogeneity in Health and Disease. Cells 2022, 11, 2021. [Google Scholar] [CrossRef]
  29. Wu, C.; Tu, T.; Xie, M.; Wang, Y.; Yan, B.; Gong, Y.; Zhang, J.; Zhou, X.; Xie, Z. Spatially resolved transcriptome of the aging mouse brain. Aging Cell 2024, 23, e14109. [Google Scholar] [CrossRef]
  30. Sun, J.; Song, Y.; Chen, Z.; Qiu, J.; Zhu, S.; Wu, L.; Xing, L. Heterogeneity and Molecular Markers for CNS Glial Cells Revealed by Single-Cell Transcriptomics. Cell. Mol. Neurobiol. 2022, 42, 2629–2642. [Google Scholar]
  31. Pettas, S.; Karagianni, K.; Kanata, E.; Chatziefstathiou, A.; Christoudia, N.; Xanthopoulos, K.; Sklaviadis, T.; Dafou, D. Profiling Microglia through Single-Cell RNA Sequencing over the Course of Development, Aging, and Disease. Cells 2022, 11, 2383. [Google Scholar] [CrossRef] [PubMed]
  32. Balak, C.D.; Han, C.Z.; Glass, C.K. Deciphering microglia phenotypes in health and disease. Curr. Opin. Genet. Dev. 2024, 84, 102146. [Google Scholar] [CrossRef] [PubMed]
  33. Qian, Z.; Qin, J.; Lai, Y.; Zhang, C.; Zhang, X. Large-Scale Integration of Single-Cell RNA-Seq Data Reveals Astrocyte Diversity and Transcriptomic Modules across Six Central Nervous System Disorders. Biomolecules 2023, 13, 692. [Google Scholar]
  34. Ahmed, S.; Gull, A.; Khuroo, T.; Aqil, M.; Sultana, Y. Glial Cell: A Potential Target for Cellular and Drug Based Therapy in Various CNS Diseases. Curr. Pharm. Des. 2017, 23, 2389–2399. [Google Scholar] [CrossRef]
  35. Liu, C.Y.; Wang, X.; Liu, C.; Zhang, H.L. Pharmacological Targeting of Microglial Activation: New Therapeutic Approach. Front. Cell. Neurosci. 2019, 13, 514. [Google Scholar] [CrossRef]
  36. Filipi, T.; Hermanova, Z.; Tureckova, J.; Vanatko, O.; Anderova, A.M. Glial Cells-The Strategic Targets in Amyotrophic Lateral Sclerosis Treatment. J. Clin. Med. 2020, 9, 261. [Google Scholar] [CrossRef]
  37. Scott, M.C.; Bedi, S.S.; Olson, S.D.; Sears, C.M.; Cox, C.S. Microglia as therapeutic targets after neurological injury: Strategy for cell therapy. Expert Opin. Ther. Targets 2021, 25, 365–380. [Google Scholar] [CrossRef]
  38. Magni, G.; Riboldi, B.; Ceruti, S. Human Glial Cells as Innovative Targets for the Therapy of Central Nervous System Pathologies. Cells 2024, 13, 606. [Google Scholar] [CrossRef]
  39. Demmings, M.D.; da Silva Chagas, L.; Traetta, M.E.; Rodrigues, R.S.; Acutain, M.F.; Barykin, E.; Datusalia, A.K.; German-Castelan, L.; Mattera, V.S.; Mazengenya, P.; et al. (Re)building the nervous system: A review of neuron-glia interactions from development to disease. J. Neurochem. 2025, 169, e16258. [Google Scholar] [CrossRef]
  40. Lannes, N.; Eppler, E.; Etemad, S.; Yotovski, P.; Filgueira, L. Microglia at center stage: A comprehensive review about the versatile and unique residential macrophages of the central nervous system. Oncotarget 2017, 8, 114393–114413. [Google Scholar] [CrossRef]
  41. Vidal-Itriago, A.; Radford, R.A.W.; Aramideh, J.A.; Maurel, C.; Scherer, N.M.; Don, E.K.; Lee, A.; Chung, R.S.; Graeber, M.B.; Morsch, M. Microglia morphophysiological diversity and its implications for the CNS. Front. Immunol. 2022, 13, 997786. [Google Scholar] [CrossRef]
  42. Qin, J.; Ma, Z.; Chen, X.; Shu, S. Microglia activation in central nervous system disorders: A review of recent mechanistic investigations and development efforts. Front. Neurol. 2023, 14, 1103416. [Google Scholar] [CrossRef] [PubMed]
  43. Biswas, K. Microglia mediated neuroinflammation in neurodegenerative diseases: A review on the cell signaling pathways involved in microglial activation. J. Neuroimmunol. 2023, 383, 578180. [Google Scholar] [CrossRef] [PubMed]
  44. Borst, K.; Dumas, A.A.; Prinz, M. Microglia: Immune and non-immune functions. Immunity 2021, 54, 2194–2208. [Google Scholar] [CrossRef]
  45. Chamkouri, H.; Motlagh Mohavi, S. Microglia and Macrophages in Central Nervous System Homeostasis and Disease Progression: Guardians and Executioners. Neuroglia 2025, 6, 31. [Google Scholar] [CrossRef]
  46. Mordelt, A.; de Witte, L.D. Microglia-mediated synaptic pruning as a key deficit in neurodevelopmental disorders: Hype or hope? Curr. Opin. Neurobiol. 2023, 79, 102674. [Google Scholar] [CrossRef]
  47. Hu, Y.; Tao, W. Current perspectives on microglia-neuron communication in the central nervous system: Direct and indirect modes of interaction. J. Adv. Res. 2024, 66, 251–265. [Google Scholar] [CrossRef]
  48. Sun, M.; You, H.; Hu, X.; Luo, Y.; Zhang, Z.; Song, Y.; An, J.; Lu, H. Microglia-Astrocyte Interaction in Neural Development and Neural Pathogenesis. Cells 2023, 12, 1942. [Google Scholar] [CrossRef]
  49. Gao, C.; Jiang, J.; Tan, Y.; Chen, S. Microglia in neurodegenerative diseases: Mechanism and potential therapeutic targets. Signal Transduct. Target. Ther. 2023, 8, 359. [Google Scholar] [CrossRef]
  50. Gradisnik, L.; Velnar, T. Astrocytes in the central nervous system and their functions in health and disease: A review. World J. Clin. Cases. 2023, 11, 3385–3394. [Google Scholar]
  51. Baldwin, K.T.; Murai, K.K.; Khakh, B.S. Astrocyte morphology. Trends Cell Biol. 2024, 34, 547–565. [Google Scholar] [CrossRef] [PubMed]
  52. Manu, D.R.; Slevin, M.; Barcutean, L.; Forro, T.; Boghitoiu, T.; Balasa, R. Astrocyte Involvement in Blood-Brain Barrier Function: A Critical Update Highlighting Novel, Complex, Neurovascular Interactions. Int. J. Mol. Sci. 2023, 24, 17146. [Google Scholar] [CrossRef] [PubMed]
  53. Theparambil, S.M.; Begum, G.; Rose, C.R. pH regulating mechanisms of astrocytes: A critical component in physiology and disease of the brain. Cell Calcium 2024, 120, 102882. [Google Scholar] [CrossRef] [PubMed]
  54. Perea, G.; Navarrete, M.; Araque, A. Tripartite synapses: Astrocytes process and control synaptic information. Trends Neurosci. 2009, 32, 421–431. [Google Scholar] [CrossRef]
  55. Harada, K.; Kamiya, T.; Tsuboi, T. Gliotransmitter Release from Astrocytes: Functional, Developmental, and Pathological Implications in the Brain. Front. Neurosci. 2016, 9, 499. [Google Scholar] [CrossRef]
  56. Kim, Y.; Dube, S.E.; Park, C.B. Brain energy homeostasis: The evolution of the astrocyte-neuron lactate shuttle hypothesis. Korean J. Physiol. Pharmacol. 2025, 29, 1–8. [Google Scholar] [CrossRef]
  57. Brambilla, R. The contribution of astrocytes to the neuroinflammatory response in multiple sclerosis and experimental autoimmune encephalomyelitis. Acta Neuropathol. 2019, 137, 757–783. [Google Scholar] [CrossRef]
  58. Markey, K.M.; Saunders, J.C.; Smuts, J.; von Reyn, C.R.; Garcia, A.D.R. Astrocyte development—More questions than answers. Front. Cell Dev. Biol. 2023, 11, 1063843. [Google Scholar] [CrossRef]
  59. Pekny, M.; Pekna, M. Astrocyte reactivity and reactive astrogliosis: Costs and benefits. Physiol. Rev. 2014, 94, 1077–1098. [Google Scholar] [CrossRef]
  60. Verkhratsky, A.; Butt, A.; Li, B.; Illes, P.; Zorec, R.; Semyanov, A.; Tang, Y.; Sofroniew, M.V. Astrocytes in human central nervous system diseases: A frontier for new therapies. Signal Transduct. Target. Ther. 2023, 8, 396. [Google Scholar] [CrossRef]
  61. Bradl, M.; Lassmann, H. Oligodendrocytes: Biology and pathology. Acta Neuropathol. 2010, 119, 37–53. [Google Scholar] [CrossRef]
  62. Kuhn, S.; Gritti, L.; Crooks, D.; Dombrowski, Y. Oligodendrocytes in Development, Myelin Generation and Beyond. Cells 2019, 8, 1424. [Google Scholar] [CrossRef]
  63. Simons, M.; Nave, K.A. Oligodendrocytes: Myelination and Axonal Support. Cold Spring Harb. Perspect. Biol. 2015, 8, a020479. [Google Scholar] [CrossRef] [PubMed]
  64. Kim, W.; Angulo, M.C. Unraveling the role of oligodendrocytes and myelin in pain. J. Neurochem. 2025, 169, e16206. [Google Scholar] [CrossRef]
  65. Zveik, O.; Rechtman, A.; Ganz, T.; Vaknin-Dembinsky, A. The interplay of inflammation and remyelination: Rethinking MS treatment with a focus on oligodendrocyte progenitor cells. Mol. Neurodegener. 2024, 19, 53. [Google Scholar] [CrossRef] [PubMed]
  66. Tepavčević, V.; Lubetzki, C. Oligodendrocyte progenitor cell recruitment and remyelination in multiple sclerosis: The more, the merrier? Brain 2022, 145, 4178–4192. [Google Scholar] [CrossRef]
  67. Molina-Gonzalez, I.; Miron, V.E.; Antel, J.P. Chronic oligodendrocyte injury in central nervous system pathologies. Commun. Biol. 2022, 5, 1274. [Google Scholar] [CrossRef] [PubMed]
  68. Liu, Y.; Yuan, J.; Dong, Y.; Jiang, S.; Zhang, M.; Zhao, X. Interaction between Oligodendrocytes and Interneurons in Brain Development and Related Neuropsychiatric Disorders. Int. J. Mol. Sci. 2024, 25, 3620. [Google Scholar] [CrossRef]
  69. García-Domínguez, M. Pathological and Inflammatory Consequences of Aging. Biomolecules 2025, 15, 404. [Google Scholar] [CrossRef]
  70. Yang, S.; Qin, C.; Hu, Z.W.; Zhou, L.Q.; Yu, H.H.; Chen, M.; Bosco, D.B.; Wang, W.; Wu, L.J.; Tian, D.S. Microglia reprogram metabolic profiles for phenotype and function changes in central nervous system. Neurobiol. Dis. 2021, 152, 105290. [Google Scholar] [CrossRef]
  71. Hoogland, I.C.; Houbolt, C.; van Westerloo, D.J.; van Gool, W.A.; van de Beek, D. Systemic inflammation and microglial activation: Systematic review of animal experiments. J. Neuroinflamm. 2015, 12, 114. [Google Scholar] [CrossRef] [PubMed]
  72. Schilling, S.; Chausse, B.; Dikmen, H.O.; Almouhanna, F.; Hollnagel, J.O.; Lewen, A.; Kann, O. TLR2- and TLR3-activated microglia induce different levels of neuronal network dysfunction in a context-dependent manner. Brain Behav. Immun. 2021, 96, 80–91. [Google Scholar] [CrossRef] [PubMed]
  73. Zhou, Y.; Chen, Y.; Xu, C.; Zhang, H.; Lin, C. TLR4 Targeting as a Promising Therapeutic Strategy for Alzheimer Disease Treatment. Front. Neurosci. 2020, 14, 602508. [Google Scholar] [CrossRef] [PubMed]
  74. Maatouk, L.; Compagnion, A.C.; Sauvage, M.C.; Bemelmans, A.P.; Leclere-Turbant, S.; Cirotteau, V.; Tohme, M.; Beke, A.; Trichet, M.; Bazin, V.; et al. TLR9 activation via microglial glucocorticoid receptors contributes to degeneration of midbrain dopamine neurons. Nat. Commun. 2018, 9, 2450. [Google Scholar] [CrossRef]
  75. Kawasaki, T.; Kawai, T. Toll-like receptor signaling pathways. Front. Immunol. 2014, 5, 461. [Google Scholar] [CrossRef]
  76. Esen, N.; Kielian, T. Central role for MyD88 in the responses of microglia to pathogen-associated molecular patterns. J. Immunol. 2006, 176, 6802–6811. [Google Scholar] [CrossRef]
  77. Anilkumar, S.; Wright-Jin, E. NF-κB as an Inducible Regulator of Inflammation in the Central Nervous System. Cells 2024, 13, 485. [Google Scholar] [CrossRef]
  78. Rodríguez-Gómez, J.A.; Kavanagh, E.; Engskog-Vlachos, P.; Engskog, M.K.R.; Herrera, A.J.; Espinosa-Oliva, A.M.; Joseph, B.; Hajji, N.; Venero, J.L.; Burguillos, M.A. Microglia: Agents of the CNS Pro-Inflammatory Response. Cells 2020, 9, 1717. [Google Scholar] [CrossRef]
  79. Li, J.; Shui, X.; Sun, R.; Wan, L.; Zhang, B.; Xiao, B.; Luo, Z. Microglial Phenotypic Transition: Signaling Pathways and Influencing Modulators Involved in Regulation in Central Nervous System Diseases. Front. Cell. Neurosci. 2021, 15, 736310. [Google Scholar] [CrossRef]
  80. Jurga, A.M.; Paleczna, M.; Kuter, K.Z. Overview of General and Discriminating Markers of Differential Microglia Phenotypes. Front. Cell. Neurosci. 2020, 14, 198. [Google Scholar] [CrossRef]
  81. Hanslik, K.L.; Ulland, T.K. The Role of Microglia and the Nlrp3 Inflammasome in Alzheimer’s Disease. Front. Neurol. 2020, 11, 570711. [Google Scholar] [CrossRef] [PubMed]
  82. Scheiblich, H.; Schlütter, A.; Golenbock, D.T.; Latz, E.; Martinez-Martinez, P.; Heneka, M.T. Activation of the NLRP3 inflammasome in microglia: The role of ceramide. J. Neurochem. 2017, 143, 534–550. [Google Scholar] [CrossRef]
  83. Lee, S.W.; de Rivero Vaccari, J.P.; Truettner, J.S.; Dietrich, W.D.; Keane, R.W. The role of microglial inflammasome activation in pyroptotic cell death following penetrating traumatic brain injury. J. Neuroinflamm. 2019, 16, 27. [Google Scholar]
  84. Wu, X.; Wan, T.; Gao, X.; Fu, M.; Duan, Y.; Shen, X.; Guo, W. Microglia Pyroptosis: A Candidate Target for Neurological Diseases Treatment. Front. Neurosci. 2022, 16, 922331. [Google Scholar] [CrossRef]
  85. Campagno, K.E.; Mitchell, C.H. The P2X7 Receptor in Microglial Cells Modulates the Endolysosomal Axis, Autophagy, and Phagocytosis. Front. Cell. Neurosci. 2021, 15, 645244. [Google Scholar] [CrossRef] [PubMed]
  86. Thawkar, B.S.; Kaur, G. Inhibitors of NF-κB and P2X7/NLRP3/Caspase 1 pathway in microglia: Novel therapeutic opportunities in neuroinflammation induced early-stage Alzheimer’s disease. J. Neuroimmunol. 2019, 326, 62–74. [Google Scholar] [CrossRef]
  87. Eyo, U.B.; Miner, S.A.; Ahlers, K.E.; Wu, L.J.; Dailey, M.E. P2X7 receptor activation regulates microglial cell death during oxygen-glucose deprivation. Neuropharmacology 2013, 73, 311–319. [Google Scholar]
  88. Anwar, S.; Pons, V.; Rivest, S. Microglia Purinoceptor P2Y6: An Emerging Therapeutic Target in CNS Diseases. Cells 2020, 9, 1595. [Google Scholar] [CrossRef]
  89. Gómez Morillas, A.; Besson, V.C.; Lerouet, D. Microglia and Neuroinflammation: What Place for P2RY12? Int. J. Mol. Sci. 2021, 22, 1636. [Google Scholar] [CrossRef]
  90. Pawelec, P.; Ziemka-Nalecz, M.; Sypecka, J.; Zalewska, T. The Impact of the CX3CL1/CX3CR1 Axis in Neurological Disorders. Cells 2020, 9, 2277. [Google Scholar]
  91. Wang, L.; Ling, H.; He, H.; Hu, N.; Xiao, L.; Zhang, Y.; Xie, L.; You, Z. Dysfunctional synaptic pruning by microglia correlates with cognitive impairment in sleep-deprived mice: Involvement of CX3CR1 signaling. Neurobiol. Stress 2023, 25, 100553. [Google Scholar] [CrossRef]
  92. Bielanin, J.P.; Sun, D. Significance of Microglial Energy Metabolism in Maintaining Brain Homeostasis. Transl. Stroke Res. 2023, 14, 435–437. [Google Scholar] [CrossRef] [PubMed]
  93. Choi, Y.K. Detrimental Roles of Hypoxia-Inducible Factor-1α in Severe Hypoxic Brain Diseases. Int. J. Mol. Sci. 2024, 25, 4465. [Google Scholar] [CrossRef]
  94. He, S.; Fan, C.; Ji, Y.; Su, Q.; Zhao, F.; Xie, C.; Chen, X.; Zhang, Y.; Chen, Y. SENP3 facilitates M1 macrophage polarization via the HIF-1α/PKM2 axis in lipopolysaccharide-induced acute lung injury. Innate Immun. 2023, 29, 25–34. [Google Scholar] [CrossRef] [PubMed]
  95. Dello Russo, C.; Lisi, L.; Tringali, G.; Navarra, P. Involvement of mTOR kinase in cytokine-dependent microglial activation and cell proliferation. Biochem. Pharmacol. 2009, 78, 1242–1251. [Google Scholar] [CrossRef] [PubMed]
  96. Tu, D.; Gao, Y.; Yang, R.; Guan, T.; Hong, J.S.; Gao, H.M. The pentose phosphate pathway regulates chronic neuroinflammation and dopaminergic neurodegeneration. J. Neuroinflamm. 2019, 16, 255. [Google Scholar] [CrossRef]
  97. Simpson, D.S.A.; Oliver, P.L. ROS Generation in Microglia: Understanding Oxidative Stress and Inflammation in Neurodegenerative Disease. Antioxidants 2020, 9, 743. [Google Scholar] [CrossRef]
  98. Kumar, A.; Chen, S.H.; Kadiiska, M.B.; Hong, J.S.; Zielonka, J.; Kalyanaraman, B.; Mason, R.P. Inducible nitric oxide synthase is key to peroxynitrite-mediated, LPS-induced protein radical formation in murine microglial BV2 cells. Free Radic. Biol. Med. 2014, 73, 51–59. [Google Scholar] [CrossRef]
  99. Ismail, F.S.; Faustmann, T.J.; Faustmann, P.M.; Corvace, F. Microglia as potential key regulators in viral-induced neuroinflammation. Front. Cell. Neurosci. 2024, 18, 1426079. [Google Scholar] [CrossRef]
  100. Kann, O.; Almouhanna, F.; Chausse, B. Interferon γ: A master cytokine in microglia-mediated neural network dysfunction and neurodegeneration. Trends Neurosci. 2022, 45, 913–927. [Google Scholar] [CrossRef]
  101. Fornari Laurindo, L.; Aparecido Dias, J.; Cressoni Araújo, A.; Torres Pomini, K.; Machado Galhardi, C.; Rucco Penteado Detregiachi, C.; Santos de Argollo Haber, L.; Donizeti Roque, D.; Dib Bechara, M.; Vialogo Marques de Castro, M.; et al. Immunological dimensions of neuroinflammation and microglial activation: Exploring innovative immunomodulatory approaches to mitigate neuroinflammatory progression. Front. Immunol. 2024, 14, 1305933. [Google Scholar] [CrossRef] [PubMed]
  102. Sofroniew, M.V. Astrocyte Reactivity: Subtypes, States, and Functions in CNS Innate Immunity. Trends Immunol. 2020, 41, 758–770. [Google Scholar] [CrossRef] [PubMed]
  103. Li, L.; Acioglu, C.; Heary, R.F.; Elkabes, S. Role of astroglial toll-like receptors (TLRs) in central nervous system infections, injury and neurodegenerative diseases. Brain Behav. Immun. 2021, 91, 740–755. [Google Scholar] [CrossRef] [PubMed]
  104. Gonzalez-Reyes, R.E.; Rubiano, M.G. Astrocyte’s RAGE: More Than Just a Question of Mood. Cent. Nerv. Syst. Agents Med. Chem. 2018, 18, 39–48. [Google Scholar] [CrossRef]
  105. Priego, N.; Zhu, L.; Monteiro, C.; Mulders, M.; Wasilewski, D.; Bindeman, W.; Doglio, L.; Martínez, L.; Martínez-Saez, E.; Ramón Cajal, S.; et al. STAT3 labels a subpopulation of reactive astrocytes required for brain metastasis. Nat. Med. 2018, 24, 1024–1035. [Google Scholar] [CrossRef]
  106. Pamies, D.; Sartori, C.; Schvartz, D.; González-Ruiz, V.; Pellerin, L.; Nunes, C.; Tavel, D.; Maillard, V.; Boccard, J.; Rudaz, S.; et al. Neuroinflammatory Response to TNFα and IL1β Cytokines Is Accompanied by an Increase in Glycolysis in Human Astrocytes In Vitro. Int. J. Mol. Sci. 2021, 22, 4065. [Google Scholar] [CrossRef]
  107. Ceyzériat, K.; Abjean, L.; Carrillo-de Sauvage, M.A.; Ben Haim, L.; Escartin, C. The complex STATes of astrocyte reactivity: How are they controlled by the JAK-STAT3 pathway? Neuroscience 2016, 330, 205–218. [Google Scholar]
  108. Villablanca, C.; Vidal, R.; Gonzalez-Billault, C. Are cytoskeleton changes observed in astrocytes functionally linked to aging? Brain Res. Bull. 2023, 196, 59–67. [Google Scholar] [CrossRef]
  109. Choi, S.S.; Lee, H.J.; Lim, I.; Satoh, J.; Kim, S.U. Human astrocytes: Secretome profiles of cytokines and chemokines. PLoS ONE 2014, 9, e92325. [Google Scholar] [CrossRef]
  110. Liu, C.Y.; Yang, Y.; Ju, W.N.; Wang, X.; Zhang, H.L. Emerging Roles of Astrocytes in Neuro-Vascular Unit and the Tripartite Synapse with Emphasis on Reactive Gliosis in the Context of Alzheimer’s Disease. Front. Cell. Neurosci. 2018, 12, 193. [Google Scholar] [CrossRef]
  111. Linnerbauer, M.; Wheeler, M.A.; Quintana, F.J. Astrocyte Crosstalk in CNS Inflammation. Neuron 2020, 108, 608–622. [Google Scholar] [CrossRef] [PubMed]
  112. Todd, A.C.; Hardingham, G.E. The Regulation of Astrocytic Glutamate Transporters in Health and Neurodegenerative Diseases. Int. J. Mol. Sci. 2020, 21, 9607. [Google Scholar] [CrossRef] [PubMed]
  113. Liddelow, S.A.; Barres, B.A. Reactive Astrocytes: Production, Function, and Therapeutic Potential. Immunity 2017, 46, 957–967. [Google Scholar] [CrossRef] [PubMed]
  114. Armada-Moreira, A.; Gomes, J.I.; Pina, C.C.; Savchak, O.K.; Gonçalves-Ribeiro, J.; Rei, N.; Pinto, S.; Morais, T.P.; Martins, R.S.; Ribeiro, F.F.; et al. Going the Extra (Synaptic) Mile: Excitotoxicity as the Road Toward Neurodegenerative Diseases. Front. Cell. Neurosci. 2020, 14, 90. [Google Scholar] [CrossRef]
  115. Verma, M.; Lizama, B.N.; Chu, C.T. Excitotoxicity, calcium and mitochondria: A triad in synaptic neurodegeneration. Transl. Neurodegener. 2022, 11, 3. [Google Scholar]
  116. Michetti, F.; D’Ambrosi, N.; Toesca, A.; Puglisi, M.A.; Serrano, A.; Marchese, E.; Corvino, V.; Geloso, M.C. The S100B story: From biomarker to active factor in neural injury. J. Neurochem. 2019, 148, 168–187. [Google Scholar]
  117. Donato, R.; Sorci, G.; Riuzzi, F.; Arcuri, C.; Bianchi, R.; Brozzi, F.; Tubaro, C.; Giambanco, I. S100B’s double life: Intracellular regulator and extracellular signal. Biochim. Biophys. Acta 2009, 1793, 1008–1022. [Google Scholar]
  118. Lee, S.; Jha, M.K.; Suk, K. Lipocalin-2 in the Inflammatory Activation of Brain Astrocytes. Crit. Rev. Immunol. 2015, 35, 77–84. [Google Scholar] [CrossRef]
  119. Tan, Q.; Zhang, C.; Rao, X.; Wan, W.; Lin, W.; Huang, S.; Ying, J.; Lin, Y.; Hua, F. The interaction of lipocalin-2 and astrocytes in neuroinflammation: Mechanisms and therapeutic application. Front. Immunol. 2024, 15, 1358719. [Google Scholar] [CrossRef]
  120. García-Domínguez, M. Relationship of S100 Proteins with Neuroinflammation. Biomolecules 2025, 15, 1125. [Google Scholar] [CrossRef]
  121. Fan, Y.Y.; Huo, J. A1/A2 astrocytes in central nervous system injuries and diseases: Angels or devils? Neurochem. Int. 2021, 148, 105080. [Google Scholar] [CrossRef]
  122. Escartin, C.; Galea, E.; Lakatos, A.; O’Callaghan, J.P.; Petzold, G.C.; Serrano-Pozo, A.; Steinhäuser, C.; Volterra, A.; Carmignoto, G.; Agarwal, A.; et al. Reactive astrocyte nomenclature, definitions, and future directions. Nat. Neurosci. 2021, 24, 312–325. [Google Scholar] [CrossRef] [PubMed]
  123. Wang, S.; Pan, Y.; Zhang, C.; Zhao, Y.; Wang, H.; Ma, H.; Sun, J.; Zhang, S.; Yao, J.; Xie, D.; et al. Transcriptome Analysis Reveals Dynamic Microglial-Induced A1 Astrocyte Reactivity via C3/C3aR/NF-κB Signaling After Ischemic Stroke. Mol. Neurobiol. 2024, 61, 10246–10270. [Google Scholar] [CrossRef] [PubMed]
  124. Chi, X.; Yin, S.; Sun, Y.; Kou, L.; Zou, W.; Wang, Y.; Jin, Z.; Wang, T.; Xia, Y. Astrocyte-neuron communication through the complement C3-C3aR pathway in Parkinson’s disease. Brain Behav. Immun. 2025, 123, 229–243. [Google Scholar] [CrossRef] [PubMed]
  125. Zhu, F.; He, P.; Jiang, W.; Afridi, S.K.; Xu, H.; Alahmad, M.; Alvin Huang, Y.W.; Qiu, W.; Wang, G.; Tang, C. Astrocyte-secreted C3 signaling impairs neuronal development and cognition in autoimmune diseases. Prog. Neurobiol. 2024, 240, 102654. [Google Scholar] [CrossRef]
  126. Wheeler, M.A.; Jaronen, M.; Covacu, R.; Zandee, S.E.J.; Scalisi, G.; Rothhammer, V.; Tjon, E.C.; Chao, C.C.; Kenison, J.E.; Blain, M.; et al. Environmental Control of Astrocyte Pathogenic Activities in CNS Inflammation. Cell 2019, 176, 581–596.e18. [Google Scholar] [CrossRef]
  127. Jha, M.K.; Jo, M.; Kim, J.H.; Suk, K. Microglia-Astrocyte Crosstalk: An Intimate Molecular Conversation. Neuroscientist 2019, 25, 227–240. [Google Scholar] [CrossRef]
  128. Miller, M.R.; Landis, H.E.; Miller, R.E.; Tizabi, Y. Intercellular Adhesion Molecule 1 (ICAM-1): An Inflammatory Regulator with Potential Implications in Ferroptosis and Parkinson’s Disease. Cells 2024, 13, 1554. [Google Scholar] [CrossRef]
  129. Shrikant, P.; Chung, I.Y.; Ballestas, M.E.; Benveniste, E.N. Regulation of intercellular adhesion molecule-1 gene expression by tumor necrosis factor-alpha, interleukin-1 beta, and interferon-gamma in astrocytes. J. Neuroimmunol. 1994, 51, 209–220. [Google Scholar]
  130. Wong, D.; Dorovini-Zis, K. Upregulation of intercellular adhesion molecule-1 (ICAM-1) expression in primary cultures of human brain microvessel endothelial cells by cytokines and lipopolysaccharide. J. Neuroimmunol. 1992, 39, 11–21. [Google Scholar] [CrossRef]
  131. Calì, C.; Cantando, I.; Veloz Castillo, M.F.; Gonzalez, L.; Bezzi, P. Metabolic Reprogramming of Astrocytes in Pathological Conditions: Implications for Neurodegenerative Diseases. Int. J. Mol. Sci. 2024, 25, 8922. [Google Scholar] [CrossRef] [PubMed]
  132. Chen, Y.; Qin, C.; Huang, J.; Tang, X.; Liu, C.; Huang, K.; Xu, J.; Guo, G.; Tong, A.; Zhou, L. The role of astrocytes in oxidative stress of central nervous system: A mixed blessing. Cell Prolif. 2020, 53, e12781. [Google Scholar] [CrossRef] [PubMed]
  133. Bokulic Panichi, L.; Stanca, S.; Dolciotti, C.; Bongioanni, P. The Role of Oligodendrocytes in Neurodegenerative Diseases: Unwrapping the Layers. Int. J. Mol. Sci. 2025, 26, 4623. [Google Scholar] [CrossRef] [PubMed]
  134. Kirby, L.; Castelo-Branco, G. Crossing boundaries: Interplay between the immune system and oligodendrocyte lineage cells. Semin. Cell Dev. Biol. 2021, 116, 45–52. [Google Scholar] [CrossRef]
  135. Emery, B.; Lu, Q.R. Transcriptional and Epigenetic Regulation of Oligodendrocyte Development and Myelination in the Central Nervous System. Cold Spring Harb. Perspect. Biol. 2015, 7, a020461. [Google Scholar] [CrossRef]
  136. Weider, M.; Starost, L.J.; Groll, K.; Küspert, M.; Sock, E.; Wedel, M.; Fröb, F.; Schmitt, C.; Baroti, T.; Hartwig, A.C.; et al. Nfat/calcineurin signaling promotes oligodendrocyte differentiation and myelination by transcription factor network tuning. Nat. Commun. 2018, 9, 899. [Google Scholar] [CrossRef]
  137. Arnett, H.A.; Mason, J.; Marino, M.; Suzuki, K.; Matsushima, G.K.; Ting, J.P. TNF alpha promotes proliferation of oligodendrocyte progenitors and remyelination. Nat. Neurosci. 2001, 4, 1116–1122. [Google Scholar] [CrossRef]
  138. Minchenberg, S.B.; Massa, P.T. The control of oligodendrocyte bioenergetics by interferon-gamma (IFN-γ) and Src homology region 2 domain-containing phosphatase-1 (SHP-1). J. Neuroimmunol. 2019, 331, 46–57. [Google Scholar] [CrossRef]
  139. Hobson, B.D.; Stanley, A.T.; De Los Santos, M.B.; Culbertson, B.; Mosharov, E.V.; Sims, P.A.; Sulzer, D. Conserved and cell type-specific transcriptional responses to IFN-γ in the ventral midbrain. Brain Behav. Immun. 2023, 111, 277–291. [Google Scholar] [CrossRef]
  140. Kirby, L.; Jin, J.; Cardona, J.G.; Smith, M.D.; Martin, K.A.; Wang, J.; Strasburger, H.; Herbst, L.; Alexis, M.; Karnell, J.; et al. Oligodendrocyte precursor cells present antigen and are cytotoxic targets in inflammatory demyelination. Nat. Commun. 2019, 10, 3887. [Google Scholar] [CrossRef]
  141. D’Souza, S.D.; Bonetti, B.; Balasingam, V.; Cashman, N.R.; Barker, P.A.; Troutt, A.B.; Raine, C.S.; Antel, J.P. Multiple sclerosis: Fas signaling in oligodendrocyte cell death. J. Exp. Med. 1996, 184, 2361–2370. [Google Scholar] [CrossRef] [PubMed]
  142. Matysiak, M.; Jurewicz, A.; Jaskolski, D.; Selmaj, K. TRAIL induces death of human oligodendrocytes isolated from adult brain. Brain 2002, 125, 2469–2480. [Google Scholar] [CrossRef]
  143. Van den Bossche, J.; Baardman, J.; Otto, N.A.; van der Velden, S.; Neele, A.E.; van den Berg, S.M.; Luque-Martin, R.; Chen, H.J.; Boshuizen, M.C.; Ahmed, M.; et al. Mitochondrial Dysfunction Prevents Repolarization of Inflammatory Macrophages. Cell Rep. 2016, 17, 684–696. [Google Scholar] [CrossRef] [PubMed]
  144. Spaas, J.; van Veggel, L.; Schepers, M.; Tiane, A.; van Horssen, J.; Wilson, D.M., 3rd; Moya, P.R.; Piccart, E.; Hellings, N.; Eijnde, B.O.; et al. Oxidative stress and impaired oligodendrocyte precursor cell differentiation in neurological disorders. Cell. Mol. Life Sci. 2021, 78, 4615–4637. [Google Scholar] [CrossRef] [PubMed]
  145. Gu, C.; Casaccia-Bonnefil, P.; Srinivasan, A.; Chao, M.V. Oligodendrocyte apoptosis mediated by caspase activation. J. Neurosci. 1999, 19, 3043–3049. [Google Scholar] [CrossRef]
  146. Druzhyna, N.M.; Musiyenko, S.I.; Wilson, G.L.; LeDoux, S.P. Cytokines induce nitric oxide-mediated mtDNA damage and apoptosis in oligodendrocytes. Protective role of targeting 8-oxoguanine glycosylase to mitochondria. J. Biol. Chem. 2005, 280, 21673–21679. [Google Scholar] [CrossRef]
  147. Wu, S.; Lin, W. The physiological role of the unfolded protein response in the nervous system. Neural Regen. Res. 2024, 19, 2411–2420. [Google Scholar] [CrossRef]
  148. Stone, S.; Wu, S.; Jamison, S.; Durose, W.; Pallais, J.P.; Lin, W. Activating transcription factor 6α deficiency exacerbates oligodendrocyte death and myelin damage in immune-mediated demyelinating diseases. Glia 2018, 66, 1331–1345. [Google Scholar] [CrossRef]
  149. Stone, S.; Lin, W. The unfolded protein response in multiple sclerosis. Front. Neurosci. 2015, 9, 264. [Google Scholar] [CrossRef]
  150. Shen, J.; Prywes, R. ER stress signaling by regulated proteolysis of ATF6. Methods 2005, 35, 382–389. [Google Scholar] [CrossRef]
  151. Lin, W.; Popko, B. Endoplasmic reticulum stress in disorders of myelinating cells. Nat. Neurosci. 2009, 12, 379–385. [Google Scholar] [CrossRef] [PubMed]
  152. Zhao, X.; Jacob, C. Mechanisms of Demyelination and Remyelination Strategies for Multiple Sclerosis. Int. J. Mol. Sci. 2023, 24, 6373. [Google Scholar] [CrossRef] [PubMed]
  153. Lopez, J.A.; Denkova, M.; Ramanathan, S.; Dale, R.C.; Brilot, F. Pathogenesis of autoimmune demyelination: From multiple sclerosis to neuromyelitis optica spectrum disorders and myelin oligodendrocyte glycoprotein antibody-associated disease. Clin. Transl. Immunol. 2021, 10, e1316. [Google Scholar] [CrossRef] [PubMed]
  154. O’Loughlin, E.; Madore, C.; Lassmann, H.; Butovsky, O. Microglial Phenotypes and Functions in Multiple Sclerosis. Cold Spring Harb. Perspect. Med. 2018, 8, a028993. [Google Scholar] [CrossRef]
  155. Krämer-Albers, E.M.; Werner, H.B. Mechanisms of axonal support by oligodendrocyte-derived extracellular vesicles. Nat. Rev. Neurosci. 2023, 24, 474–486. [Google Scholar] [CrossRef]
  156. Zajicek, J.; Wing, M.; Skepper, J.; Compston, A. Human oligodendrocytes are not sensitive to complement. A study of CD59 expression in the human central nervous system. Lab. Investig. 1995, 73, 128–138. [Google Scholar]
  157. Soane, L.; Cho, H.J.; Niculescu, F.; Rus, H.; Shin, M.L. C5b-9 terminal complement complex protects oligodendrocytes from death by regulating Bad through phosphatidylinositol 3-kinase/Akt pathway. J. Immunol. 2001, 167, 2305–2311. [Google Scholar] [CrossRef]
  158. Negro-Demontel, L.; Maleki, A.F.; Reich, D.S.; Kemper, C. The complement system in neurodegenerative and inflammatory diseases of the central nervous system. Front. Neurol. 2024, 15, 1396520. [Google Scholar] [CrossRef]
  159. Xu, X.; Yang, M.; Zhang, B.; Dong, J.; Zhuang, Y.; Ge, Q.; Niu, F.; Liu, B. HIF-1α participates in secondary brain injury through regulating neuroinflammation. Transl. Neurosci. 2023, 14, 20220272. [Google Scholar] [CrossRef]
  160. Yang, C.; Pan, R.Y.; Guan, F.; Yuan, Z. Lactate metabolism in neurodegenerative diseases. Neural Regen. Res. 2024, 19, 69–74. [Google Scholar] [CrossRef]
  161. Garland, E.F.; Hartnell, I.J.; Boche, D. Microglia and Astrocyte Function and Communication: What Do We Know in Humans? Front. Neurosci. 2022, 16, 824888. [Google Scholar] [CrossRef] [PubMed]
  162. Molnár, T.; Héja, L.; Emri, Z.; Simon, A.; Nyitrai, G.; Pál, I.; Kardos, J. Activation of astroglial calcium signaling by endogenous metabolites succinate and gamma-hydroxybutyrate in the nucleus accumbens. Front. Neuroenergetics 2011, 3, 7. [Google Scholar] [CrossRef] [PubMed]
  163. Domercq, M.; Vázquez-Villoldo, N.; Matute, C. Neurotransmitter signaling in the pathophysiology of microglia. Front. Cell. Neurosci. 2013, 7, 49. [Google Scholar] [PubMed]
  164. Li, H.; Zhao, Y.; Dai, R.; Geng, P.; Weng, D.; Wu, W.; Yu, F.; Lin, R.; Wu, Z.; Li, Y.; et al. Astrocytes release ATP/ADP and glutamate in flashes via vesicular exocytosis. Mol. Psychiatry 2025, 30, 2475–2489. [Google Scholar]
  165. D’Souza, S.D.; Antel, J.P.; Freedman, M.S. Cytokine induction of heat shock protein expression in human oligodendrocytes: An interleukin-1-mediated mechanism. J. Neuroimmunol. 1994, 50, 17–24. [Google Scholar] [CrossRef]
  166. Peferoen, L.; Kipp, M.; van der Valk, P.; van Noort, J.M.; Amor, S. Oligodendrocyte-microglia cross-talk in the central nervous system. Immunology 2014, 141, 302–313. [Google Scholar] [CrossRef]
  167. Teleanu, D.M.; Niculescu, A.G.; Lungu, I.I.; Radu, C.I.; Vladâcenco, O.; Roza, E.; Costăchescu, B.; Grumezescu, A.M.; Teleanu, R.I. An Overview of Oxidative Stress, Neuroinflammation, and Neurodegenerative Diseases. Int. J. Mol. Sci. 2022, 23, 5938. [Google Scholar] [CrossRef]
  168. Chamani, S.; Bianconi, V.; Tasbandi, A.; Pirro, M.; Barreto, G.E.; Jamialahmadi, T.; Sahebkar, A. Resolution of Inflammation in Neurodegenerative Diseases: The Role of Resolvins. Mediat. Inflamm. 2020, 2020, 3267172. [Google Scholar] [CrossRef]
  169. Müller, L.; Di Benedetto, S. Neuroimmune crosstalk in chronic neuroinflammation: Microglial interactions and immune modulation. Front. Cell. Neurosci. 2025, 19, 1575022. [Google Scholar] [CrossRef]
  170. Yu, Y.; Chen, R.; Mao, K.; Deng, M.; Li, Z. The Role of Glial Cells in Synaptic Dysfunction: Insights into Alzheimer’s Disease Mechanisms. Aging Dis. 2024, 15, 459–479. [Google Scholar]
  171. Soteros, B.M.; Sia, G.M. Complement and microglia dependent synapse elimination in brain development. WIREs Mech. Dis. 2022, 14, e1545. [Google Scholar] [CrossRef]
  172. Gomez-Arboledas, A.; Acharya, M.M.; Tenner, A.J. The Role of Complement in Synaptic Pruning and Neurodegeneration. Immunotargets Ther. 2021, 10, 373–386. [Google Scholar] [CrossRef] [PubMed]
  173. Geloso, M.C.; D’Ambrosi, N. Microglial Pruning: Relevance for Synaptic Dysfunction in Multiple Sclerosis and Related Experimental Models. Cells 2021, 10, 686. [Google Scholar] [CrossRef] [PubMed]
  174. Chung, W.S.; Clarke, L.E.; Wang, G.X.; Stafford, B.K.; Sher, A.; Chakraborty, C.; Joung, J.; Foo, L.C.; Thompson, A.; Chen, C.; et al. Astrocytes mediate synapse elimination through MEGF10 and MERTK pathways. Nature 2013, 504, 394–400. [Google Scholar] [CrossRef] [PubMed]
  175. Zabel, M.K.; Kirsch, W.M. From development to dysfunction: Microglia and the complement cascade in CNS homeostasis. Ageing Res. Rev. 2013, 12, 749–756. [Google Scholar] [CrossRef]
  176. Crehan, H.; Hardy, J.; Pocock, J. Microglia, Alzheimer’s disease, and complement. Int. J. Alzheimers Dis. 2012, 2012, 983640. [Google Scholar] [CrossRef]
  177. Hughes, A.N.; Appel, B. Microglia phagocytose myelin sheaths to modify developmental myelination. Nat. Neurosci. 2020, 23, 1055–1066. [Google Scholar] [CrossRef]
  178. Zrzavy, T.; Hametner, S.; Wimmer, I.; Butovsky, O.; Weiner, H.L.; Lassmann, H. Loss of ‘homeostatic’ microglia and patterns of their activation in active multiple sclerosis. Brain 2017, 140, 1900–1913. [Google Scholar] [CrossRef]
  179. Henstridge, C.M.; Pickett, E.; Spires-Jones, T.L. Synaptic pathology: A shared mechanism in neurological disease. Ageing Res. Rev. 2016, 28, 72–84. [Google Scholar] [CrossRef]
  180. Lecca, D.; Jung, Y.J.; Scerba, M.T.; Hwang, I.; Kim, Y.K.; Kim, S.; Modrow, S.; Tweedie, D.; Hsueh, S.C.; Liu, D.; et al. Role of chronic neuroinflammation in neuroplasticity and cognitive function: A hypothesis. Alzheimers Dement. 2022, 18, 2327–2340. [Google Scholar] [CrossRef]
  181. Distéfano-Gagné, F.; Bitarafan, S.; Lacroix, S.; Gosselin, D. Roles and regulation of microglia activity in multiple sclerosis: Insights from animal models. Nat. Rev. Neurosci. 2023, 24, 397–415. [Google Scholar] [CrossRef] [PubMed]
  182. Olcum, M.; Tastan, B.; Kiser, C.; Genc, S.; Genc, K. Microglial NLRP3 inflammasome activation in multiple sclerosis. Adv. Protein Chem. Struct. Biol. 2020, 119, 247–308. [Google Scholar] [PubMed]
  183. Chastain, E.M.; Duncan, D.S.; Rodgers, J.M.; Miller, S.D. The role of antigen presenting cells in multiple sclerosis. Biochim. Biophys. Acta 2011, 1812, 265–274. [Google Scholar] [PubMed]
  184. Abulaban, A.A.; Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Albuhadily, A.K.; Shokr, M.M.; Alexiou, A.; Papadakis, M.; Batiha, G.E. The janus face of astrocytes in multiple sclerosis: Balancing protection and pathology. Brain Res. Bull. 2025, 226, 111356. [Google Scholar] [CrossRef]
  185. Kostic, M.; Zivkovic, N.; Stojanovic, I. Multiple sclerosis and glutamate excitotoxicity. Rev. Neurosci. 2013, 24, 71–88. [Google Scholar]
  186. García-Domínguez, M. White Matter in Crisis: Oligodendrocytes and the Pathophysiology of Multiple Sclerosis. Cells 2025, 14, 1408. [Google Scholar] [CrossRef]
  187. Kim, Y.S.; Jung, H.M.; Yoon, B.E. Exploring glia to better understand Alzheimer’s disease. Anim. Cells Syst. 2018, 22, 213–218. [Google Scholar] [CrossRef]
  188. Leng, F.; Edison, P. Neuroinflammation and microglial activation in Alzheimer disease: Where do we go from here? Nat. Rev. Neurol. 2021, 17, 157–172. [Google Scholar]
  189. Kim, J.; Yoo, I.D.; Lim, J.; Moon, J.S. Pathological phenotypes of astrocytes in Alzheimer’s disease. Exp. Mol. Med. 2024, 56, 95–99. [Google Scholar] [CrossRef]
  190. Yeung, J.H.Y.; Palpagama, T.H.; Wood, O.W.G.; Turner, C.; Waldvogel, H.J.; Faull, R.L.M.; Kwakowsky, A. EAAT2 Expression in the Hippocampus, Subiculum, Entorhinal Cortex and Superior Temporal Gyrus in Alzheimer’s Disease. Front. Cell. Neurosci. 2021, 15, 702824. [Google Scholar] [CrossRef]
  191. Yang, X.; Chen, Y.H.; Liu, L.; Gu, Z.; You, Y.; Hao, J.R.; Sun, N.; Gao, C. Regulation of glycolysis-derived L-lactate production in astrocytes rescues the memory deficits and Aβ burden in early Alzheimer’s disease models. Pharmacol. Res. 2024, 208, 107357. [Google Scholar] [CrossRef] [PubMed]
  192. Liao, B.; Zhao, W.; Beers, D.R.; Henkel, J.S.; Appel, S.H. Transformation from a neuroprotective to a neurotoxic microglial phenotype in a mouse model of ALS. Exp. Neurol. 2012, 237, 147–152. [Google Scholar] [CrossRef] [PubMed]
  193. Brites, D.; Vaz, A.R. Microglia centered pathogenesis in ALS: Insights in cell interconnectivity. Front. Cell. Neurosci. 2014, 8, 117. [Google Scholar] [CrossRef] [PubMed]
  194. Provenzano, F.; Torazza, C.; Bonifacino, T.; Bonanno, G.; Milanese, M. The Key Role of Astrocytes in Amyotrophic Lateral Sclerosis and Their Commitment to Glutamate Excitotoxicity. Int. J. Mol. Sci. 2023, 24, 15430. [Google Scholar] [CrossRef]
  195. Jamet, M.; Dupuis, L.; Gonzalez De Aguilar, J.L. Oligodendrocytes in amyotrophic lateral sclerosis and frontotemporal dementia: The new players on stage. Front. Mol. Neurosci. 2024, 17, 1375330. [Google Scholar] [CrossRef]
  196. Ho, M.S. Microglia in Parkinson’s Disease. Adv. Exp. Med. Biol. 2019, 1175, 335–353. [Google Scholar]
  197. Dustin, C.M.; Shiva, S.S.; Vazquez, A.; Saeed, A.; Pascoal, T.; Cifuentes-Pagano, E.; Pagano, P.J. NOX2 in Alzheimer’s and Parkinson’s disease. Redox Biol. 2024, 78, 103433. [Google Scholar] [CrossRef]
  198. Garcia, R.; Zarate, S.; Srinivasan, R. The Role of Astrocytes in Parkinson’s Disease: Astrocytes in Parkinson’s Disease. Adv. Neurobiol. 2024, 39, 319–343. [Google Scholar]
  199. Miyazaki, I.; Asanuma, M. Neuron-Astrocyte Interactions in Parkinson’s Disease. Cells 2020, 9, 2623. [Google Scholar] [CrossRef]
  200. Lista, S.; Imbimbo, B.P.; Grasso, M.; Fidilio, A.; Emanuele, E.; Minoretti, P.; López-Ortiz, S.; Martín-Hernández, J.; Gabelle, A.; Caruso, G.; et al. Tracking neuroinflammatory biomarkers in Alzheimer’s disease: A strategy for individualized therapeutic approaches? J. Neuroinflamm. 2024, 21, 187. [Google Scholar] [CrossRef]
  201. Thomas, G.; Rahman, R. Evolution of Preclinical Models for Glioblastoma Modelling and Drug Screening. Curr. Oncol. Rep. 2025, 27, 601–624. [Google Scholar] [CrossRef] [PubMed]
  202. Gilron, I.; Xiao, M.Z.X.; Carley, M.; Salter, M.W.; Hutchinson, M.R.; Moulin, D.E.; Moore, R.A.; Ross-White, A. Glial-modulating agents for the treatment of pain: A systematic review. Pain 2025, 166, 1030–1049. [Google Scholar] [CrossRef] [PubMed]
  203. Wawrzyniak, A.; Krawczyk-Marć, I.; Żuryń, A.; Walocha, J.; Balawender, K. Diversity, Functional Complexity, and Translational Potential of Glial Cells in the Central Nervous System. Int. J. Mol. Sci. 2025, 26, 9080. [Google Scholar] [CrossRef] [PubMed]
  204. Tsau, S.; Emerson, M.R.; Lynch, S.G.; LeVine, S.M. Aspirin and multiple sclerosis. BMC Med. 2015, 13, 153. [Google Scholar] [CrossRef]
  205. Rivers-Auty, J.; Mather, A.E.; Peters, R.; Lawrence, C.B.; Brough, D. Anti-inflammatories in Alzheimer’s disease-potential therapy or spurious correlate? Brain Commun. 2020, 2, fcaa109. [Google Scholar] [CrossRef]
  206. Ding, P.; Gorenflo, M.P.; Zhu, X.; Xu, R. Aspirin Use and Risk of Alzheimer’s Disease: A 2-Sample Mendelian Randomization Study. J. Alzheimers Dis. 2023, 92, 989–1000. [Google Scholar] [CrossRef]
  207. Gao, X.; Chen, H.; Schwarzschild, M.A.; Ascherio, A. Use of ibuprofen and risk of Parkinson disease. Neurology 2011, 76, 863–869. [Google Scholar] [CrossRef]
  208. Badawoud, A.M.; Ali, L.S.; Abdallah, M.S.; El Sabaa, R.M.; Bahaa, M.M.; Elmasry, T.A.; Wahsh, E.; Yasser, M.; Eltantawy, N.; Eldesoqui, M.; et al. The relation between Parkinson’s disease and non-steroidal anti-inflammatories; a systematic review and meta-analysis. Front. Pharmacol. 2024, 15, 1434512. [Google Scholar] [CrossRef]
  209. Khrieba, M.O.; Hegazy, S.K.; Mustafa, W.; El-Haggar, S.M. Repurposing celecoxib as adjuvant therapy in patients with Parkinsonian disease: A new therapeutic dawn: Randomized controlled pilot study. Inflammopharmacology 2024, 32, 3729–3738. [Google Scholar] [CrossRef]
  210. Bhanja, D.; Hallan, D.R.; Staub, J.; Rizk, E.; Zacko, J.C. Early Celecoxib use in Patients with Traumatic Brain Injury. Neurocrit. Care 2024, 40, 886–897. [Google Scholar] [CrossRef]
  211. Tsai, C.P.; Lin, F.C.; Lee, J.K.; Lee, C.T. Aspirin use associated with amyotrophic lateral sclerosis: A total population-based case-control study. J. Epidemiol. 2015, 25, 172–177. [Google Scholar] [CrossRef]
  212. Salomon-Zimri, S.; Pushett, A.; Russek-Blum, N.; Van Eijk, R.P.A.; Birman, N.; Abramovich, B.; Eitan, E.; Elgrart, K.; Beaulieu, D.; Ennist, D.L.; et al. Combination of ciprofloxacin/celecoxib as a novel therapeutic strategy for ALS. Amyotroph. Lateral Scler. Front. Degener. 2023, 24, 263–271. [Google Scholar] [CrossRef]
  213. Hawkey, C.J. COX-1 and COX-2 inhibitors. Best. Pract. Res. Clin. Gastroenterol. 2001, 15, 801–820. [Google Scholar] [CrossRef] [PubMed]
  214. Kaufmann, W.E.; Andreasson, K.I.; Isakson, P.C.; Worley, P.F. Cyclooxygenases and the central nervous system. Prostaglandins 1997, 54, 601–624. [Google Scholar] [CrossRef] [PubMed]
  215. Minghetti, L. Cyclooxygenase-2 (COX-2) in inflammatory and degenerative brain diseases. J. Neuropathol. Exp. Neurol. 2004, 63, 901–910. [Google Scholar] [CrossRef] [PubMed]
  216. Dembo, G.; Park, S.B.; Kharasch, E.D. Central nervous system concentrations of cyclooxygenase-2 inhibitors in humans. Anesthesiology 2005, 102, 409–415. [Google Scholar] [CrossRef]
  217. Cheng, L.; Hu, Z.; Gu, J.; Li, Q.; Liu, J.; Liu, M.; Li, J.; Bi, X. Exploring COX-Independent Pathways: A Novel Approach for Meloxicam and Other NSAIDs in Cancer and Cardiovascular Disease Treatment. Pharmaceuticals 2024, 17, 1488. [Google Scholar] [CrossRef]
  218. Puhl, A.C.; Milton, F.A.; Cvoro, A.; Sieglaff, D.H.; Campos, J.C.; Bernardes, A.; Filgueira, C.S.; Lindemann, J.L.; Deng, T.; Neves, F.A.; et al. Mechanisms of peroxisome proliferator activated receptor γ regulation by non-steroidal anti-inflammatory drugs. Nucl. Recept. Signal. 2015, 13, e004. [Google Scholar] [CrossRef]
  219. Machado, M.M.F.; Bassani, T.B.; Cóppola-Segovia, V.; Moura, E.L.R.; Zanata, S.M.; Andreatini, R.; Vital, M.A.B.F. PPAR-γ agonist pioglitazone reduces microglial proliferation and NF-κB activation in the substantia nigra in the 6-hydroxydopamine model of Parkinson’s disease. Pharmacol. Rep. 2019, 71, 556–564. [Google Scholar] [CrossRef]
  220. Mannila, A.; Rautio, J.; Lehtonen, M.; Järvinen, T.; Savolainen, J. Inefficient central nervous system delivery limits the use of ibuprofen in neurodegenerative diseases. Eur. J. Pharm. Sci. 2005, 24, 101–105. [Google Scholar] [CrossRef]
  221. Stiller, C.O.; Hjemdahl, P. Lessons from 20 years with COX-2 inhibitors: Importance of dose-response considerations and fair play in comparative trials. J. Intern. Med. 2022, 292, 557–574. [Google Scholar] [CrossRef]
  222. Aisen, P.S. Evaluation of selective COX-2 inhibitors for the treatment of Alzheimer’s disease. J. Pain Symptom Manage. 2002, 23, S35–S40. [Google Scholar] [CrossRef] [PubMed]
  223. Rashid, M.A.; Tang, J.J.; Yoo, K.H.; Corujo-Ramirez, A.; Oliveros, A.; Kim, S.H.; Ullah, F.; Altawell, R.; Hawse, J.R.; Cole, P.D.; et al. The selective cyclooxygenase-2 inhibitor NS398 ameliorates cisplatin-induced impairments in mitochondrial and cognitive function. Front. Mol. Neurosci. 2023, 16, 1295991. [Google Scholar] [CrossRef] [PubMed]
  224. Harirforoosh, S.; Asghar, W.; Jamali, F. Adverse effects of nonsteroidal antiinflammatory drugs: An update of gastrointestinal, cardiovascular and renal complications. J. Pharm. Pharm. Sci. 2013, 16, 821–847. [Google Scholar] [CrossRef] [PubMed]
  225. Brogden, R.N.; Speight, T.M.; Avery, G.S. Minocycline: A review of its antibacterial and pharmacokinetic properties and therapeutic use. Drugs 1975, 9, 251–291. [Google Scholar] [CrossRef]
  226. Plane, J.M.; Shen, Y.; Pleasure, D.E.; Deng, W. Prospects for minocycline neuroprotection. Arch. Neurol. 2010, 67, 1442–1448. [Google Scholar] [CrossRef]
  227. Abraham, J.; Fox, P.D.; Condello, C.; Bartolini, A.; Koh, S. Minocycline attenuates microglia activation and blocks the long-term epileptogenic effects of early-life seizures. Neurobiol. Dis. 2012, 46, 425–430. [Google Scholar] [CrossRef]
  228. Zimmerman, A.J.; Mangano, N.; Park, G.; Kaushal, A.K.; Bergese, S.D. Glial Modulator Antibiotics for Neuropathic Pain: Current Insights and Future Directions. Pharmaceuticals 2025, 18, 346. [Google Scholar] [CrossRef]
  229. Zhu, S.; Stavrovskaya, I.G.; Drozda, M.; Kim, B.Y.; Ona, V.; Li, M.; Sarang, S.; Liu, A.S.; Hartley, D.M.; Wu, D.C.; et al. Minocycline inhibits cytochrome c release and delays progression of amyotrophic lateral sclerosis in mice. Nature 2002, 417, 74–78. [Google Scholar] [CrossRef]
  230. Dominic, M.R. Adverse Reactions Induced by Minocycline: A Review of Literature. Curr. Drug Saf. 2021, 16, 309–321. [Google Scholar] [CrossRef]
  231. Coisne, C.; Mao, W.; Engelhardt, B. Cutting edge: Natalizumab blocks adhesion but not initial contact of human T cells to the blood-brain barrier in vivo in an animal model of multiple sclerosis. J. Immunol. 2009, 182, 5909–5913. [Google Scholar] [CrossRef] [PubMed]
  232. Sucksdorff, M.; Tuisku, J.; Matilainen, M.; Vuorimaa, A.; Smith, S.; Keitilä, J.; Rokka, J.; Parkkola, R.; Nylund, M.; Rinne, J.; et al. Natalizumab treatment reduces microglial activation in the white matter of the MS brain. Neurol. Neuroimmunol. Neuroinflamm. 2019, 6, e574. [Google Scholar] [CrossRef] [PubMed]
  233. Li, H.; Shi, F.H.; Huang, S.Y.; Zhang, S.G.; Gu, Z.C.; Wei, J.F. Clinical adverse effects of natalizumab: Protocol for a meta-analysis of randomized double-blind placebo-controlled clinical trails. Medicine 2018, 97, e11507. [Google Scholar] [CrossRef] [PubMed]
  234. Liu, Y.; Liu, S.; Liu, L.; Gong, X.; Liu, J.; Sun, L.; Liu, X.; Wu, L.; Chen, L.; Wang, L.; et al. Fine Comparison of the Efficacy and Safety Between GB242 and Infliximab in Patients with Rheumatoid Arthritis: A Phase III Study. Rheumatol. Ther. 2022, 9, 175–189. [Google Scholar] [CrossRef]
  235. Traczewski, P.; Rudnicka, L. Adalimumab in dermatology. Br. J. Clin. Pharmacol. 2008, 66, 618–625. [Google Scholar] [CrossRef]
  236. Dadsetan, S.; Balzano, T.; Forteza, J.; Agusti, A.; Cabrera-Pastor, A.; Taoro-Gonzalez, L.; Hernandez-Rabaza, V.; Gomez-Gimenez, B.; ElMlili, N.; Llansola, M.; et al. Infliximab reduces peripheral inflammation, neuroinflammation, and extracellular GABA in the cerebellum and improves learning and motor coordination in rats with hepatic encephalopathy. J. Neuroinflamm. 2016, 13, 245. [Google Scholar] [CrossRef]
  237. Strangfeld, A.; Listing, J. Infection and musculoskeletal conditions: Bacterial and opportunistic infections during anti-TNF therapy. Best Pract. Res. Clin. Rheumatol. 2006, 20, 1181–1195. [Google Scholar]
  238. Kim, S.Y.; Solomon, D.H. Tumor necrosis factor blockade and the risk of viral infection. Nat. Rev. Rheumatol. 2010, 6, 165–174. [Google Scholar] [CrossRef]
  239. Li, Y.; Ye, R.; Dai, H.; Lin, J.; Cheng, Y.; Zhou, Y.; Lu, Y. Exploring TNFR1: From discovery to targeted therapy development. J. Transl. Med. 2025, 23, 71. [Google Scholar] [CrossRef]
  240. Shivaji, U.N.; Sharratt, C.L.; Thomas, T.; Smith, S.C.L.; Iacucci, M.; Moran, G.W.; Ghosh, S.; Bhala, N. Review article: Managing the adverse events caused by anti-TNF therapy in inflammatory bowel disease. Aliment. Pharmacol. Ther. 2019, 49, 664–680. [Google Scholar]
  241. Torene, R.; Nirmala, N.; Obici, L.; Cattalini, M.; Tormey, V.; Caorsi, R.; Starck-Schwertz, S.; Letzkus, M.; Hartmann, N.; Abrams, K.; et al. Canakinumab reverses overexpression of inflammatory response genes in tumour necrosis factor receptor-associated periodic syndrome. Ann. Rheum. Dis. 2017, 76, 303–309. [Google Scholar] [CrossRef]
  242. Thompson, P.L.; Nidorf, S.M. Anti-inflammatory therapy with canakinumab for atherosclerotic disease: Lessons from the CANTOS trial. J. Thorac. Dis. 2018, 10, 695–698. [Google Scholar] [CrossRef] [PubMed]
  243. Weickert, T.W.; Jacomb, I.; Lenroot, R.; Lappin, J.; Weinberg, D.; Brooks, W.S.; Brown, D.; Pellen, D.; Kindler, J.; Mohan, A.; et al. Adjunctive canakinumab reduces peripheral inflammation markers and improves positive symptoms in people with schizophrenia and inflammation: A randomized control trial. Brain Behav. Immun. 2024, 115, 191–200. [Google Scholar] [CrossRef] [PubMed]
  244. Kuemmerle-Deschner, J.B.; Kallinich, T.; Henes, J.; Kortus-Götze, B.; Oommen, P.T.; Rech, J.; Krickau, T.; Weller-Heinemann, F.; Horneff, G.; Janda, A.; et al. Long-term safety and effectiveness of canakinumab in patients with monogenic autoinflammatory diseases: Results from the interim analysis of the RELIANCE registry. RMD Open 2024, 10, e003890. [Google Scholar] [CrossRef] [PubMed]
  245. Parisi, S.; Ditto, M.C.; Ghellere, F.; Panaro, S.; Piccione, F.; Borrelli, R.; Fusaro, E. Update on tocilizumab in rheumatoid arthritis: A narrative review. Front. Immunol. 2025, 16, 1470488. [Google Scholar] [CrossRef]
  246. Yu, Y.M.; Jin, G.H.; Zhong, C.; Qian, H.; Wang, L.; Zhan, F. Exploring the role of interleukin-6 receptor blockade in epilepsy and associated neuropsychiatric conditions through a mendelian randomization study. World J. Psychiatry 2024, 14, 1244–1253. [Google Scholar] [CrossRef]
  247. Sevigny, J.; Chiao, P.; Bussière, T.; Weinreb, P.H.; Williams, L.; Maier, M.; Dunstan, R.; Salloway, S.; Chen, T.; Ling, Y.; et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature 2016, 537, 50–56. [Google Scholar] [CrossRef]
  248. Cadiz, M.P.; Gibson, K.A.; Todd, K.T.; Nascari, D.G.; Massa, N.; Lilley, M.T.; Olney, K.C.; Al-Amin, M.M.; Jiang, H.; Holtzman, D.M.; et al. Aducanumab anti-amyloid immunotherapy induces sustained microglial and immune alterations. J. Exp. Med. 2024, 221, e20231363. [Google Scholar] [CrossRef]
  249. Nisticò, R.; Borg, J.J. Aducanumab for Alzheimer’s disease: A regulatory perspective. Pharmacol. Res. 2021, 171, 105754. [Google Scholar] [CrossRef]
  250. Hu, H.; Zhao, Y.; Mao, J.; He, J.; Zhang, Y.; Ye, H.; Yang, X. A real-world pharmacovigilance study of adverse drug reactions associated with lecanemab and aducanumab based on WHO-VigiAccess and FAERS databases. Front. Pharmacol. 2025, 16, 1561020. [Google Scholar] [CrossRef]
  251. Yrjänheikki, J.; Tikka, T.; Keinänen, R.; Goldsteins, G.; Chan, P.H.; Koistinaho, J. A tetracycline derivative, minocycline, reduces inflammation and protects against focal cerebral ischemia with a wide therapeutic window. Proc. Natl. Acad. Sci. USA 1999, 96, 13496–13500. [Google Scholar] [CrossRef]
  252. Wells, J.E.; Hurlbert, R.J.; Fehlings, M.G.; Yong, V.W. Neuroprotection by minocycline facilitates significant recovery from spinal cord injury in mice. Brain 2003, 126, 1628–1637. [Google Scholar] [CrossRef] [PubMed]
  253. Brundula, V.; Rewcastle, N.B.; Metz, L.M.; Bernard, C.C.; Yong, V.W. Targeting leukocyte MMPs and transmigration: Minocycline as a potential therapy for multiple sclerosis. Brain 2002, 125, 1297–1308. [Google Scholar] [CrossRef] [PubMed]
  254. Stirling, D.P.; Khodarahmi, K.; Liu, J.; McPhail, L.T.; McBride, C.B.; Steeves, J.D.; Ramer, M.S.; Tetzlaff, W. Minocycline treatment reduces delayed oligodendrocyte death, attenuates axonal dieback, and improves functional outcome after spinal cord injury. J. Neurosci. 2004, 24, 2182–2190. [Google Scholar] [CrossRef] [PubMed]
  255. Carris, N.W.; Pardo, J.; Montero, J.; Shaeer, K.M. Minocycline as A Substitute for Doxycycline in Targeted Scenarios: A Systematic Review. Open Forum Infect. Dis. 2015, 2, ofv178. [Google Scholar] [CrossRef]
  256. Kothari, M.; Wanjari, A.; Acharya, S.; Karwa, V.; Chavhan, R.; Kumar, S.; Kadu, A.; Patil, R. A Comprehensive Review of Monoclonal Antibodies in Modern Medicine: Tracing the Evolution of a Revolutionary Therapeutic Approach. Cureus 2024, 16, e61983. [Google Scholar] [CrossRef]
  257. Fernandez-Bueno, I.; Garcia-Gutierrez, M.T.; Srivastava, G.K.; Gayoso, M.J.; Gonzalo-Orden, J.M.; Pastor, J.C. Adalimumab (tumor necrosis factor-blocker) reduces the expression of glial fibrillary acidic protein immunoreactivity increased by exogenous tumor necrosis factor alpha in an organotypic culture of porcine neuroretina. Mol. Vis. 2013, 19, 894–903. [Google Scholar]
  258. Sirbu, C.A.; Budisteanu, M.; Falup-Pecurariu, C. Monoclonal antibodies—A revolutionary therapy in multiple sclerosis. Neurol. Neurochir. Pol. 2020, 54, 21–27. [Google Scholar] [CrossRef]
  259. Hu, B.; Duan, S.; Wang, Z.; Li, X.; Zhou, Y.; Zhang, X.; Zhang, Y.W.; Xu, H.; Zheng, H. Insights Into the Role of CSF1R in the Central Nervous System and Neurological Disorders. Front. Aging Neurosci. 2021, 13, 789834. [Google Scholar] [CrossRef]
  260. Stanley, E.R.; Chitu, V. CSF-1 receptor signaling in myeloid cells. Cold Spring Harb. Perspect. Biol. 2014, 6, a021857. [Google Scholar] [CrossRef]
  261. Okojie, A.K.; Uweru, J.O.; Coburn, M.A.; Li, S.; Cao-Dao, V.D.; Eyo, U.B. Distinguishing the effects of systemic CSF1R inhibition by PLX3397 on microglia and peripheral immune cells. J. Neuroinflamm. 2023, 20, 242. [Google Scholar] [CrossRef] [PubMed]
  262. Neal, M.L.; Fleming, S.M.; Budge, K.M.; Boyle, A.M.; Kim, C.; Alam, G.; Beier, E.E.; Wu, L.J.; Richardson, J.R. Pharmacological inhibition of CSF1R by GW2580 reduces microglial proliferation and is protective against neuroinflammation and dopaminergic neurodegeneration. FASEB J. 2020, 34, 1679–1694. [Google Scholar] [CrossRef] [PubMed]
  263. Sosna, J.; Philipp, S.; Albay, R., 3rd; Reyes-Ruiz, J.M.; Baglietto-Vargas, D.; LaFerla, F.M.; Glabe, C.G. Early long-term administration of the CSF1R inhibitor PLX3397 ablates microglia and reduces accumulation of intraneuronal amyloid, neuritic plaque deposition and pre-fibrillar oligomers in 5XFAD mouse model of Alzheimer’s disease. Mol. Neurodegener. 2018, 13, 11. [Google Scholar] [CrossRef] [PubMed]
  264. Oh, S.J.; Ahn, H.; Jung, K.H.; Han, S.J.; Nam, K.R.; Kang, K.J.; Park, J.A.; Lee, K.C.; Lee, Y.J.; Choi, J.Y. Evaluation of the Neuroprotective Effect of Microglial Depletion by CSF-1R Inhibition in a Parkinson’s Animal Model. Mol. Imaging Biol. 2020, 22, 1031–1042. [Google Scholar] [CrossRef]
  265. Olmos-Alonso, A.; Schetters, S.T.; Sri, S.; Askew, K.; Mancuso, R.; Vargas-Caballero, M.; Holscher, C.; Perry, V.H.; Gomez-Nicola, D. Pharmacological targeting of CSF1R inhibits microglial proliferation and prevents the progression of Alzheimer’s-like pathology. Brain 2016, 139, 891–907. [Google Scholar]
  266. León-Rodríguez, A.; Grondona, J.M.; Marín-Wong, S.; López-Aranda, M.F.; López-Ávalos, M.D. Long-term reprogramming of primed microglia after moderate inhibition of CSF1R signaling. Glia 2025, 73, 175–195. [Google Scholar] [CrossRef]
  267. Pocock, J.; Vasilopoulou, F.; Svensson, E.; Cosker, K. Microglia and TREM2. Neuropharmacology 2024, 257, 110020. [Google Scholar] [CrossRef]
  268. Deczkowska, A.; Weiner, A.; Amit, I. The Physiology, Pathology, and Potential Therapeutic Applications of the TREM2 Signaling Pathway. Cell 2020, 181, 1207–1217. [Google Scholar] [CrossRef]
  269. Deczkowska, A.; Keren-Shaul, H.; Weiner, A.; Colonna, M.; Schwartz, M.; Amit, I. Disease-Associated Microglia: A Universal Immune Sensor of Neurodegeneration. Cell 2018, 173, 1073–1081. [Google Scholar] [CrossRef]
  270. Long, H.; Simmons, A.; Mayorga, A.; Burgess, B.; Nguyen, T.; Budda, B.; Rychkova, A.; Rhinn, H.; Tassi, I.; Ward, M.; et al. Preclinical and first-in-human evaluation of AL002, a novel TREM2 agonistic antibody for Alzheimer’s disease. Alzheimers Res. Ther. 2024, 16, 235. [Google Scholar] [CrossRef]
  271. Tisavipat, N.; Juan, H.Y.; Chen, J.J. Monoclonal antibody therapies for aquaporin-4-immunoglobulin G-positive neuromyelitis optica spectrum disorder and myelin oligodendrocyte glycoprotein antibody-associated disease. Saudi J. Ophthalmol. 2023, 38, 2–12. [Google Scholar] [CrossRef] [PubMed]
  272. Tradtrantip, L.; Zhang, H.; Anderson, M.O.; Saadoun, S.; Phuan, P.W.; Papadopoulos, M.C.; Bennett, J.L.; Verkman, A.S. Small-molecule inhibitors of NMO-IgG binding to aquaporin-4 reduce astrocyte cytotoxicity in neuromyelitis optica. FASEB J. 2012, 26, 2197–2208. [Google Scholar] [CrossRef] [PubMed]
  273. Okada, M.; Oka, T.; Nakamoto, M.; Fukuyama, K.; Shiroyama, T. Astroglial Connexin43 as a Potential Target for a Mood Stabiliser. Int. J. Mol. Sci. 2020, 22, 339. [Google Scholar] [CrossRef] [PubMed]
  274. Tonkin, R.S.; Bowles, C.; Perera, C.J.; Keating, B.A.; Makker, P.G.S.; Duffy, S.S.; Lees, J.G.; Tran, C.; Don, A.S.; Fath, T.; et al. Attenuation of mechanical pain hypersensitivity by treatment with Peptide5, a connexin-43 mimetic peptide, involves inhibition of NLRP3 inflammasome in nerve-injured mice. Exp. Neurol. 2018, 300, 1–12. [Google Scholar] [CrossRef]
  275. Xiong, X.; Chen, W.; Chen, C.; Wu, Q.; He, C. Analysis of the function and therapeutic strategy of connexin 43 from its subcellular localization. Biochimie 2024, 218, 1–7. [Google Scholar] [CrossRef]
  276. Melzer, N.; Meuth, S.G.; Torres-Salazar, D.; Bittner, S.; Zozulya, A.L.; Weidenfeller, C.; Kotsiari, A.; Stangel, M.; Fahlke, C.; Wiendl, H. A beta-lactam antibiotic dampens excitotoxic inflammatory CNS damage in a mouse model of multiple sclerosis. PLoS ONE 2008, 3, e3149. [Google Scholar] [CrossRef]
  277. Limpert, A.S.; Cosford, N.D. Translational enhancers of EAAT2: Therapeutic implications for neurodegenerative disease. J. Clin. Investig. 2014, 124, 964–967. [Google Scholar] [CrossRef]
  278. Rotolo, R.A.; Demuro, J.; Drummond, G.; Little, C.; Johns, L.D.; Betz, A.J. Prophylactic exposure to oral riluzole reduces the clinical severity and immune-related biomarkers of experimental autoimmune encephalomyelitis. J. Neuroimmunol. 2021, 356, 577603. [Google Scholar] [CrossRef]
  279. Kalafatakis, I.; Papagianni, F.; Theodorakis, K.; Karagogeos, D. Nogo-A and LINGO-1: Two Important Targets for Remyelination and Regeneration. Int. J. Mol. Sci. 2023, 24, 4479. [Google Scholar] [CrossRef]
  280. Eidsvaag, V.A.; Enger, R.; Hansson, H.A.; Eide, P.K.; Nagelhus, E.A. Human and mouse cortical astrocytes differ in aquaporin-4 polarization toward microvessels. Glia 2017, 65, 964–973. [Google Scholar] [CrossRef]
  281. Gundersen, G.A.; Vindedal, G.F.; Skare, O.; Nagelhus, E.A. Evidence that pericytes regulate aquaporin-4 polarization in mouse cortical astrocytes. Brain Struct. Funct. 2014, 219, 2181–2186. [Google Scholar] [CrossRef] [PubMed]
  282. Salman, M.M.; Kitchen, P.; Halsey, A.; Wang, M.X.; Törnroth-Horsefield, S.; Conner, A.C.; Badaut, J.; Iliff, J.J.; Bill, R.M. Emerging roles for dynamic aquaporin-4 subcellular relocalization in CNS water homeostasis. Brain 2022, 145, 64–75. [Google Scholar] [CrossRef] [PubMed]
  283. Nesverova, V.; Törnroth-Horsefield, S. Phosphorylation-Dependent Regulation of Mammalian Aquaporins. Cells 2019, 8, 82. [Google Scholar] [CrossRef] [PubMed]
  284. Fukuda, A.M.; Badaut, J. Aquaporin 4: A player in cerebral edema and neuroinflammation. J. Neuroinflamm. 2012, 9, 279. [Google Scholar] [CrossRef]
  285. Vandebroek, A.; Yasui, M. Regulation of AQP4 in the Central Nervous System. Int. J. Mol. Sci. 2020, 21, 1603. [Google Scholar] [CrossRef]
  286. Lawrence, J.M.; Schardien, K.; Wigdahl, B.; Nonnemacher, M.R. Roles of neuropathology-associated reactive astrocytes: A systematic review. Acta Neuropathol. Commun. 2023, 11, 42. [Google Scholar] [CrossRef]
  287. Huang, X.; Su, Y.; Wang, N.; Li, H.; Li, Z.; Yin, G.; Chen, H.; Niu, J.; Yi, C. Astroglial Connexins in Neurodegenerative Diseases. Front. Mol. Neurosci. 2021, 14, 657514. [Google Scholar] [CrossRef]
  288. Márquez-Rosado, L.; Solan, J.L.; Dunn, C.A.; Norris, R.P.; Lampe, P.D. Connexin43 phosphorylation in brain, cardiac, endothelial and epithelial tissues. Biochim. Biophys. Acta 2012, 1818, 1985–1992. [Google Scholar] [CrossRef]
  289. Chen, Y.; Wang, L.; Zhang, L.; Chen, B.; Yang, L.; Li, X.; Li, Y.; Yu, H. Inhibition of Connexin 43 Hemichannels Alleviates Cerebral Ischemia/Reperfusion Injury via the TLR4 Signaling Pathway. Front. Cell. Neurosci. 2018, 12, 372. [Google Scholar] [CrossRef]
  290. Pajarillo, E.; Rizor, A.; Lee, J.; Aschner, M.; Lee, E. The role of astrocytic glutamate transporters GLT-1 and GLAST in neurological disorders: Potential targets for neurotherapeutics. Neuropharmacology 2019, 161, 107559. [Google Scholar] [CrossRef]
  291. Grewer, C.; Rauen, T. Electrogenic glutamate transporters in the CNS: Molecular mechanism, pre-steady-state kinetics, and their impact on synaptic signaling. J. Membr. Biol. 2005, 203, 1–20. [Google Scholar] [CrossRef] [PubMed]
  292. Karki, P.; Webb, A.; Smith, K.; Lee, K.; Son, D.S.; Aschner, M.; Lee, E. cAMP response element-binding protein (CREB) and nuclear factor κB mediate the tamoxifen-induced up-regulation of glutamate transporter 1 (GLT-1) in rat astrocytes. J. Biol. Chem. 2013, 288, 28975–28986. [Google Scholar] [CrossRef] [PubMed]
  293. Green, J.L.; Dos Santos, W.F.; Fontana, A.C.K. Role of glutamate excitotoxicity and glutamate transporter EAAT2 in epilepsy: Opportunities for novel therapeutics development. Biochem. Pharmacol. 2021, 193, 114786. [Google Scholar] [CrossRef] [PubMed]
  294. Howland, D.S.; Liu, J.; She, Y.; Goad, B.; Maragakis, N.J.; Kim, B.; Erickson, J.; Kulik, J.; DeVito, L.; Psaltis, G.; et al. Focal loss of the glutamate transporter EAAT2 in a transgenic rat model of SOD1 mutant-mediated amyotrophic lateral sclerosis (ALS). Proc. Natl. Acad. Sci. USA 2002, 99, 1604–1609. [Google Scholar] [CrossRef]
  295. Wood, O.W.; Yeung, J.H.; Kwakowsky, A. Is EAAT2 a potential therapeutic intervention target for Alzheimer’s disease? Neural Regen. Res. 2023, 18, 1721–1722. [Google Scholar]
  296. Bhatnagar, A.; Parmar, V.; Barbieri, N.; Bearoff, F.; Elefant, F.; Kortagere, S. Novel EAAT2 activators improve motor and cognitive impairment in a transgenic model of Huntington’s disease. Front. Behav. Neurosci. 2023, 17, 1176777. [Google Scholar] [CrossRef]
  297. Alijanpour, S.; Miryounesi, M.; Ghafouri-Fard, S. The role of excitatory amino acid transporter 2 (EAAT2) in epilepsy and other neurological disorders. Metab. Brain Dis. 2023, 38, 1–16. [Google Scholar] [CrossRef]
  298. Rashidbenam, Z.; Ozturk, E.; Pagnin, M.; Theotokis, P.; Grigoriadis, N.; Petratos, S. How does Nogo receptor influence demyelination and remyelination in the context of multiple sclerosis? Front. Cell. Neurosci. 2023, 17, 1197492. [Google Scholar] [CrossRef]
  299. Andrews, J.L.; Fernandez-Enright, F. A decade from discovery to therapy: Lingo-1, the dark horse in neurological and psychiatric disorders. Neurosci. Biobehav. Rev. 2015, 56, 97–114. [Google Scholar] [CrossRef]
  300. Jha, M.K.; Morrison, B.M. Lactate Transporters Mediate Glia-Neuron Metabolic Crosstalk in Homeostasis and Disease. Front. Cell. Neurosci. 2020, 14, 589582. [Google Scholar] [CrossRef]
  301. Tepavčević, V. Oligodendroglial Energy Metabolism and (re)Myelination. Life 2021, 11, 238. [Google Scholar] [CrossRef]
  302. Lee, Y.; Morrison, B.M.; Li, Y.; Lengacher, S.; Farah, M.H.; Hoffman, P.N.; Liu, Y.; Tsingalia, A.; Jin, L.; Zhang, P.W.; et al. Oligodendroglia metabolically support axons and contribute to neurodegeneration. Nature 2012, 487, 443–448. [Google Scholar] [CrossRef] [PubMed]
  303. Shi, C.; Xu, J.; Ding, Y.; Chen, X.; Yuan, F.; Zhu, F.; Duan, C.; Hu, J.; Lu, H.; Wu, T.; et al. MCT1-mediated endothelial cell lactate shuttle as a target for promoting axon regeneration after spinal cord injury. Theranostics 2024, 14, 5662–5681. [Google Scholar] [CrossRef] [PubMed]
  304. Chohan, M.O. Deconstructing Neurogenesis, Transplantation and Genome-Editing as Neural Repair Strategies in Brain Disease. Front. Cell Dev. Biol. 2020, 8, 116. [Google Scholar] [CrossRef] [PubMed]
  305. Wang, J.H.; Gessler, D.J.; Zhan, W.; Gallagher, T.L.; Gao, G. Adeno-associated virus as a delivery vector for gene therapy of human diseases. Signal Transduct. Target. Ther. 2024, 9, 78. [Google Scholar] [CrossRef]
  306. O’Carroll, S.J.; Cook, W.H.; Young, D. AAV Targeting of Glial Cell Types in the Central and Peripheral Nervous System and Relevance to Human Gene Therapy. Front. Mol. Neurosci. 2021, 13, 618020. [Google Scholar] [CrossRef]
  307. Serrano, C.; Cananzi, S.; Shen, T.; Wang, L.L.; Zhang, C.L. Simple and highly specific targeting of resident microglia with adeno-associated virus. iScience 2024, 27, 110706. [Google Scholar] [CrossRef]
  308. Dodge, J.C.; Haidet, A.M.; Yang, W.; Passini, M.A.; Hester, M.; Clarke, J.; Roskelley, E.M.; Treleaven, C.M.; Rizo, L.; Martin, H.; et al. Delivery of AAV-IGF-1 to the CNS extends survival in ALS mice through modification of aberrant glial cell activity. Mol. Ther. 2008, 16, 1056–1064. [Google Scholar] [CrossRef]
  309. Henry, R.A.; Hughes, S.M.; Connor, B. AAV-mediated delivery of BDNF augments neurogenesis in the normal and quinolinic acid-lesioned adult rat brain. Eur. J. Neurosci. 2007, 25, 3513–3525. [Google Scholar] [CrossRef]
  310. Zhou, L.; Wang, Y.; Xu, Y.; Zhang, Y.; Zhu, C. A comprehensive review of AAV-mediated strategies targeting microglia for therapeutic intervention of neurodegenerative diseases. J. Neuroinflamm. 2024, 21, 232. [Google Scholar] [CrossRef]
  311. Yudaeva, A.; Kostyusheva, A.; Kachanov, A.; Brezgin, S.; Ponomareva, N.; Parodi, A.; Pokrovsky, V.S.; Lukashev, A.; Chulanov, V.; Kostyushev, D. Clinical and Translational Landscape of Viral Gene Therapies. Cells 2024, 13, 1916. [Google Scholar] [CrossRef]
  312. Dhuri, K.; Bechtold, C.; Quijano, E.; Pham, H.; Gupta, A.; Vikram, A.; Bahal, R. Antisense Oligonucleotides: An Emerging Area in Drug Discovery and Development. J. Clin. Med. 2020, 9, 2004. [Google Scholar] [CrossRef] [PubMed]
  313. Zhang, J.; Chen, B.; Gan, C.; Sun, H.; Zhang, J.; Feng, L. A Comprehensive Review of Small Interfering RNAs (siRNAs): Mechanism, Therapeutic Targets, and Delivery Strategies for Cancer Therapy. Int. J. Nanomed. 2023, 18, 7605–7635. [Google Scholar] [CrossRef]
  314. Iyer, A.K.; Schoch, K.M.; Verbeck, A.; Galasso, G.; Chen, H.; Smith, S.; Oldenborg, A.; Miller, T.M.; Karch, C.M.; Bonni, A. Targeted ASO-mediated Atp1a2 knockdown in astrocytes reduces SOD1 aggregation and accelerates disease onset in mutant SOD1 mice. PLoS ONE 2023, 18, e0294731. [Google Scholar] [CrossRef] [PubMed]
  315. Chong, Z.Z.; Souayah, N. Targeting Gene C9orf72 Pathogenesis for Amyotrophic Lateral Sclerosis. Int. J. Mol. Sci. 2025, 26, 4276. [Google Scholar] [CrossRef] [PubMed]
  316. Min, H.S.; Kim, H.J.; Naito, M.; Ogura, S.; Toh, K.; Hayashi, K.; Kim, B.S.; Fukushima, S.; Anraku, Y.; Miyata, K.; et al. Systemic Brain Delivery of Antisense Oligonucleotides across the Blood-Brain Barrier with a Glucose-Coated Polymeric Nanocarrier. Angew. Chem. Int. Ed. Engl. 2020, 59, 8173–8180. [Google Scholar]
  317. Shao, Q.; Zhang, Y.; Zhang, Z.; Jiang, W.; Yin, Y.; Fang, Y.; Zhang, C.; Chen, Q.; Ning, B. Downregulation of Circular RNA Gla Reduced Astrocyte Inflammatory Status by Regulating miR-488/MEKK1 Levels and Promoted Functional Recovery After Spinal Cord Injury. J. Inflamm. Res. 2024, 17, 7123–7139. [Google Scholar] [CrossRef]
  318. Zheng, X.; Wang, W. Astrocyte transplantation for repairing the injured spinal cord. J. Biomed. Res. 2022, 36, 312–320. [Google Scholar] [CrossRef]
  319. Lepore, A.C.; Rauck, B.; Dejea, C.; Pardo, A.C.; Rao, M.S.; Rothstein, J.D.; Maragakis, N.J. Focal transplantation-based astrocyte replacement is neuroprotective in a model of motor neuron disease. Nat. Neurosci. 2008, 11, 1294–1301. [Google Scholar] [CrossRef]
  320. Davies, S.J.; Shih, C.H.; Noble, M.; Mayer-Proschel, M.; Davies, J.E.; Proschel, C. Transplantation of specific human astrocytes promotes functional recovery after spinal cord injury. PLoS ONE 2011, 6, e17328. [Google Scholar] [CrossRef]
  321. Hastings, N.; Kuan, W.L.; Osborne, A.; Kotter, M.R.N. Therapeutic Potential of Astrocyte Transplantation. Cell Transplant. 2022, 31, 9636897221105499. [Google Scholar] [CrossRef] [PubMed]
  322. Zheng, Y.; Gallegos, C.M.; Xue, H.; Li, S.; Kim, D.H.; Zhou, H.; Xia, X.; Liu, Y.; Cao, Q. Transplantation of Human Induced Pluripotent Stem Cell-Derived Neural Progenitor Cells Promotes Forelimb Functional Recovery after Cervical Spinal Cord Injury. Cells 2022, 11, 2765. [Google Scholar] [CrossRef] [PubMed]
  323. Tchieu, J.; Calder, E.L.; Guttikonda, S.R.; Gutzwiller, E.M.; Aromolaran, K.A.; Steinbeck, J.A.; Goldstein, P.A.; Studer, L. NFIA is a gliogenic switch enabling rapid derivation of functional human astrocytes from pluripotent stem cells. Nat. Biotechnol. 2019, 37, 267–275. [Google Scholar] [CrossRef] [PubMed]
  324. Li, N.; Leung, G.K. Oligodendrocyte Precursor Cells in Spinal Cord Injury: A Review and Update. Biomed. Res. Int. 2015, 2015, 235195. [Google Scholar] [CrossRef]
  325. Wu, B.; Sun, L.; Li, P.; Tian, M.; Luo, Y.; Ren, X. Transplantation of oligodendrocyte precursor cells improves myelination and promotes functional recovery after spinal cord injury. Injury 2012, 43, 794–801. [Google Scholar] [CrossRef]
  326. Sun, Y.; Xu, C.C.; Li, J.; Guan, X.Y.; Gao, L.; Ma, L.X.; Li, R.X.; Peng, Y.W.; Zhu, G.P. Transplantation of oligodendrocyte precursor cells improves locomotion deficits in rats with spinal cord irradiation injury. PLoS ONE 2013, 8, e57534. [Google Scholar] [CrossRef]
  327. Keirstead, H.S.; Nistor, G.; Bernal, G.; Totoiu, M.; Cloutier, F.; Sharp, K.; Steward, O. Human embryonic stem cell-derived oligodendrocyte progenitor cell transplants remyelinate and restore locomotion after spinal cord injury. J. Neurosci. 2005, 25, 4694–4705. [Google Scholar] [CrossRef]
  328. Wang, S.; Bates, J.; Li, X.; Schanz, S.; Chandler-Militello, D.; Levine, C.; Maherali, N.; Studer, L.; Hochedlinger, K.; Windrem, M.; et al. Human iPSC-derived oligodendrocyte progenitor cells can myelinate and rescue a mouse model of congenital hypomyelination. Cell Stem Cell 2013, 12, 252–264. [Google Scholar] [CrossRef]
  329. Zhou, K.; Wang, Y.; Xu, Y.; Zhang, Y.; Zhu, C. Exploring microglial replacement: From disease models to clinical translation. Brain Behav. Immun. 2025, 129, 179–185. [Google Scholar] [CrossRef]
  330. Wu, J.; Wang, Y.; Li, X.; Ouyang, P.; Cai, Y.; He, Y.; Zhang, M.; Luan, X.; Jin, Y.; Wang, J.; et al. Microglia replacement halts the progression of microgliopathy in mice and humans. Science 2025, 389, eadr1015. [Google Scholar] [CrossRef]
  331. Mader, M.M.; Scavetti, A.; Yoo, Y.; Chai, A.T.; Uenaka, T.; Wernig, M. Therapeutic genetic restoration through allogeneic brain microglia replacement. Nature 2025, 646, 903–912. [Google Scholar] [CrossRef]
  332. Frosch, M.; Shimizu, T.; Wogram, E.; Amann, L.; Gruber, L.; Groisman, A.I.; Fliegauf, M.; Schwabenland, M.; Chhatbar, C.; Zechel, S.; et al. Microglia-neuron crosstalk via Hex-GM2-MGL2 maintains brain homeostasis. Nature 2025, 8086, 913–924. [Google Scholar] [CrossRef]
  333. Aisenberg, W.H.; O’Brien, C.A.; Sangster, M.; Yaqoob, F.; Zhang, Y.; Temsamrit, B.; Thom, S.; Gosse, L.; Chaluvadi, S.; Elfayomi, B.; et al. Direct microglia replacement reveals pathologic and therapeutic contributions of brain macrophages to a monogenic neurological disease. Immunity 2025, 58, 1254–1268.e9. [Google Scholar] [CrossRef] [PubMed]
  334. Serneels, L.; Sierksma, A.; Pasciuto, E.; Geric, I.; Nair, A.; Martinez-Muriana, A.; Snellinx, A.; De Strooper, B. A versatile mouse model to advance human microglia transplantation research in neurodegenerative diseases. Mol. Neurodegener. 2025, 20, 29. [Google Scholar] [CrossRef] [PubMed]
  335. Sweeney, M.D.; Zhao, Z.; Montagne, A.; Nelson, A.R.; Zlokovic, B.V. Blood-Brain Barrier: From Physiology to Disease and Back. Physiol. Rev. 2019, 99, 21–78. [Google Scholar] [CrossRef] [PubMed]
  336. Saraiva, C.; Praça, C.; Ferreira, R.; Santos, T.; Ferreira, L.; Bernardino, L. Nanoparticle-mediated brain drug delivery: Overcoming blood-brain barrier to treat neurodegenerative diseases. J. Control. Release 2016, 235, 34–47. [Google Scholar] [CrossRef]
  337. Chen, Y.; Liu, L. Modern methods for delivery of drugs across the blood-brain barrier. Adv. Drug Deliv. Rev. 2012, 64, 640–665. [Google Scholar] [CrossRef]
  338. Ahlawat, J.; Guillama Barroso, G.; Masoudi Asil, S.; Alvarado, M.; Armendariz, I.; Bernal, J.; Carabaza, X.; Chavez, S.; Cruz, P.; Escalante, V.; et al. Nanocarriers as Potential Drug Delivery Candidates for Overcoming the Blood-Brain Barrier: Challenges and Possibilities. ACS Omega 2020, 5, 12583–12595. [Google Scholar] [CrossRef]
  339. Zhou, Z.; Sun, T.; Jiang, C. Recent advances on drug delivery nanocarriers for cerebral disorders. Biomed. Mater. 2021, 16, 024104. [Google Scholar] [CrossRef]
  340. Thomsen, M.S.; Johnsen, K.B.; Kucharz, K.; Lauritzen, M.; Moos, T. Blood-Brain Barrier Transport of Transferrin Receptor-Targeted Nanoparticles. Pharmaceutics 2022, 14, 2237. [Google Scholar] [CrossRef]
  341. Fu, C.; Xiang, Y.; Li, X.; Fu, A. Targeted transport of nanocarriers into brain for theranosis with rabies virus glycoprotein-derived peptide. Mater. Sci. Eng. C. Mater. Biol. Appl. 2018, 87, 155–166. [Google Scholar] [CrossRef] [PubMed]
  342. Xiao, H.; Amarsaikhan, O.; Zhao, Y.; Yu, X.; Hu, X.; Han, S.; Chaolumen; Baigude, H. Astrocyte-targeted siRNA delivery by adenosine-functionalized LNP in mouse TBI model. Mol. Ther. Nucleic Acids 2023, 34, 102065. [Google Scholar] [CrossRef] [PubMed]
  343. Zhao, N.; Francis, N.L.; Calvelli, H.R.; Moghe, P.V. Microglia-targeting nanotherapeutics for neurodegenerative diseases. APL Bioeng. 2020, 4, 030902. [Google Scholar] [CrossRef]
  344. Lei, S.Y.; Yang, Y.Q.; Liu, J.C.; Zhang, D.H.; Qu, Y.; Sun, Y.Y.; Zhu, H.J.; Zhou, S.Y.; Yang, Y.; Guo, Z.N. Nanodrug delivery systems for regulating microglial polarization in ischemic stroke treatment: A review. J. Tissue Eng. 2024, 15, 20417314241237052. [Google Scholar] [CrossRef] [PubMed]
  345. Miao, J.; Chen, L.; Pan, X.; Li, L.; Zhao, B.; Lan, J. Microglial Metabolic Reprogramming: Emerging Insights and Therapeutic Strategies in Neurodegenerative Diseases. Cell. Mol. Neurobiol. 2023, 43, 3191–3210. [Google Scholar] [CrossRef]
  346. Banks, W.A.; Sharma, P.; Bullock, K.M.; Hansen, K.M.; Ludwig, N.; Whiteside, T.L. Transport of Extracellular Vesicles across the Blood-Brain Barrier: Brain Pharmacokinetics and Effects of Inflammation. Int. J. Mol. Sci. 2020, 21, 4407. [Google Scholar] [CrossRef]
  347. Wiklander, O.P.B.; Brennan, M.Á.; Lötvall, J.; Breakefield, X.O.; El Andaloussi, S. Advances in therapeutic applications of extracellular vesicles. Sci. Transl. Med. 2019, 11, eaav8521. [Google Scholar] [CrossRef]
  348. Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345. [Google Scholar] [CrossRef]
  349. Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 2019, 9, 19. [Google Scholar] [CrossRef]
  350. McLaughlin, C.; Datta, P.; Singh, Y.P.; Lo, A.; Horchler, S.; Elcheva, I.A.; Ozbolat, I.T.; Ravnic, D.J.; Koduru, S.V. Mesenchymal Stem Cell-Derived Extracellular Vesicles for Therapeutic Use and in Bioengineering Applications. Cells 2022, 11, 3366. [Google Scholar] [CrossRef]
  351. Mavroudis, I.; Balmus, I.M.; Ciobica, A.; Nicoara, M.N.; Luca, A.C.; Palade, D.O. The Role of Microglial Exosomes and miR-124-3p in Neuroinflammation and Neuronal Repair after Traumatic Brain Injury. Life 2023, 13, 1924. [Google Scholar] [CrossRef]
  352. Liu, W.; Bai, X.; Zhang, A.; Huang, J.; Xu, S.; Zhang, J. Role of Exosomes in Central Nervous System Diseases. Front. Mol. Neurosci. 2019, 12, 240. [Google Scholar] [CrossRef]
  353. Grimm, H.P.; Schumacher, V.; Schäfer, M.; Imhof-Jung, S.; Freskgård, P.O.; Brady, K.; Hofmann, C.; Rüger, P.; Schlothauer, T.; Göpfert, U.; et al. Delivery of the Brainshuttle™ amyloid-beta antibody fusion trontinemab to non-human primate brain and projected efficacious dose regimens in humans. mAbs 2023, 15, 2261509. [Google Scholar] [CrossRef]
  354. Jin, B.K.; Odongo, S.; Radwanska, M.; Magez, S. NANOBODIES®: A Review of Diagnostic and Therapeutic Applications. Int. J. Mol. Sci. 2023, 24, 5994. [Google Scholar] [CrossRef]
  355. Zheng, F.; Pang, Y.; Li, L.; Pang, Y.; Zhang, J.; Wang, X.; Raes, G. Applications of nanobodies in brain diseases. Front. Immunol. 2022, 13, 978513. [Google Scholar] [CrossRef]
  356. Zhu, K.; Wang, Y.; Sarlus, H.; Geng, K.; Nutma, E.; Sun, J.; Kung, S.Y.; Bay, C.; Han, J.; Min, J.H.; et al. Myeloid cell-specific topoisomerase 1 inhibition using DNA origami mitigates neuroinflammation. EMBO Rep. 2022, 23, e54499. [Google Scholar] [CrossRef]
Figure 1. This image illustrates the process of remyelination in the CNS through three sequential stages. The loss of myelin sheaths caused by neuronal injury during demyelination first accumulates myelin debris, which is then removed by macrophages and/or microglial cells. Microglial cells secrete several soluble factors that promote OPCs differentiation into mature pre-myelinating oligodendrocytes in the second stage. These differentiated oligodendrocytes ultimately regenerate the myelin sheaths surrounding axons, thus reestablishing proper neuronal function during the remyelination phase. In demyelinating disorders such as MS, this process is critical for the restoration of neural integrity and functional recovery. Abbreviations: OPC (oligodendrocyte precursor cell).
Figure 1. This image illustrates the process of remyelination in the CNS through three sequential stages. The loss of myelin sheaths caused by neuronal injury during demyelination first accumulates myelin debris, which is then removed by macrophages and/or microglial cells. Microglial cells secrete several soluble factors that promote OPCs differentiation into mature pre-myelinating oligodendrocytes in the second stage. These differentiated oligodendrocytes ultimately regenerate the myelin sheaths surrounding axons, thus reestablishing proper neuronal function during the remyelination phase. In demyelinating disorders such as MS, this process is critical for the restoration of neural integrity and functional recovery. Abbreviations: OPC (oligodendrocyte precursor cell).
Biomedicines 14 00115 g001
Figure 2. Microglial polarization and its impact on neuronal outcomes. Resting microglial cells maintains homeostasis through surveillance functions mediated by molecules like CSF1R, SIRP1A, CX3CL1, and CD200R. Upon activation, microglial cells can polarize into either a pro-inflammatory (M1) or anti-inflammatory (M2) phenotype. M1 microglial cells are induced by classical activation signals such as PAMPs (e.g., LPS), IFN-γ, and GM-CSF, secrete several pro-inflammatory mediators including IL-1β, IL-6, IL-12, TNF-α, CCL2, CXCL10, and iNOS, and express MHC-II, CD86, and CD16/32, leading to neurotoxic effects and neuronal degeneration. In contrast, alternative activation by IL-4, IL-10, or IgG promotes an M2 phenotype characterized by anti-inflammatory and neuroprotective responses, including the production of IL-10, TGF-β, and expression of CD206, SOCS3, Arg1, Fizz1, and Ym1, supporting neuronal survival and maintaining healthy neurons. The balance between M1 and M2 phenotypes determines the outcome of neuroinflammation and is critical in neurodegenerative and demyelinating diseases. Abbreviations: PAMP (pathogen-associated molecular pattern), LPS (lipopolysaccharide), IFN-γ (interferon gamma), GM-CSF (granulocyte-macrophage colony-stimulating factor), IL-4 (interleukin 4), IL-10 (interleukin 10), IgG (immunoglobulin G), IL-1β (interleukin 1 beta), IL-6 (interleukin 6), IL-12 (interleukin 12), TNF-α (tumor necrosis factor alpha), CCL2 (C-C motif chemokine ligand 2), CXCL10 (C-X-C motif chemokine ligand 10), iNOS (inducible nitric oxide synthase), MHC-II (major histocompatibility complex class II), CD86 (cluster of differentiation 86), CD16 (cluster of differentiation 16), CD32 (cluster of differentiation 32), IL-10 (interleukin-10), TGF-β (transforming growth factor beta), CD206 (cluster of differentiation 206), SOCS3 (suppressor of cytokine signaling 3), Arg1 (arginase 1), Fizz1 (found in inflammatory zone 1), Ym1 (chitinase-like protein 3), CSF1R (colony-stimulating factor 1 receptor), SIRP1A (signal regulatory protein alpha), CX3CL1 (C-X3-C motif chemokine ligand 1), and CD200R (CD200 receptor).
Figure 2. Microglial polarization and its impact on neuronal outcomes. Resting microglial cells maintains homeostasis through surveillance functions mediated by molecules like CSF1R, SIRP1A, CX3CL1, and CD200R. Upon activation, microglial cells can polarize into either a pro-inflammatory (M1) or anti-inflammatory (M2) phenotype. M1 microglial cells are induced by classical activation signals such as PAMPs (e.g., LPS), IFN-γ, and GM-CSF, secrete several pro-inflammatory mediators including IL-1β, IL-6, IL-12, TNF-α, CCL2, CXCL10, and iNOS, and express MHC-II, CD86, and CD16/32, leading to neurotoxic effects and neuronal degeneration. In contrast, alternative activation by IL-4, IL-10, or IgG promotes an M2 phenotype characterized by anti-inflammatory and neuroprotective responses, including the production of IL-10, TGF-β, and expression of CD206, SOCS3, Arg1, Fizz1, and Ym1, supporting neuronal survival and maintaining healthy neurons. The balance between M1 and M2 phenotypes determines the outcome of neuroinflammation and is critical in neurodegenerative and demyelinating diseases. Abbreviations: PAMP (pathogen-associated molecular pattern), LPS (lipopolysaccharide), IFN-γ (interferon gamma), GM-CSF (granulocyte-macrophage colony-stimulating factor), IL-4 (interleukin 4), IL-10 (interleukin 10), IgG (immunoglobulin G), IL-1β (interleukin 1 beta), IL-6 (interleukin 6), IL-12 (interleukin 12), TNF-α (tumor necrosis factor alpha), CCL2 (C-C motif chemokine ligand 2), CXCL10 (C-X-C motif chemokine ligand 10), iNOS (inducible nitric oxide synthase), MHC-II (major histocompatibility complex class II), CD86 (cluster of differentiation 86), CD16 (cluster of differentiation 16), CD32 (cluster of differentiation 32), IL-10 (interleukin-10), TGF-β (transforming growth factor beta), CD206 (cluster of differentiation 206), SOCS3 (suppressor of cytokine signaling 3), Arg1 (arginase 1), Fizz1 (found in inflammatory zone 1), Ym1 (chitinase-like protein 3), CSF1R (colony-stimulating factor 1 receptor), SIRP1A (signal regulatory protein alpha), CX3CL1 (C-X3-C motif chemokine ligand 1), and CD200R (CD200 receptor).
Biomedicines 14 00115 g002
Figure 3. Diagram illustrating the mechanism of synaptic pruning in the CNS. Synaptic pruning is a key process in the developing and adult brain where excess or weak synapses are eliminated to refine neural circuits. In this mechanism, glutamate is released from the presynaptic terminal and binds to NMDA and AMPA receptors on the postsynaptic neuron, facilitating neurotransmission. Astrocytes maintain synaptic homeostasis by clearing excess glutamate via the GLT1 transporter. Meanwhile, signals like TGF-β from the presynaptic terminal induce the expression and relocalization of complement protein C1q, which, along with C3, tags synapses for elimination. These tags are recognized by CR3/CR4 receptors on microglial cells, which then engulf the marked synapses. Astrocytes also contribute to synaptic pruning through phagocytic receptors such as MERTK and MEGF10, regulated by the STAT3-SOCS3 pathway. Moreover, IL-33 released through astroglial end-feet further activates microglial pruning. The coordinated interplay of neurons, astrocytes, and microglial cells regulates synaptic remodeling during development, with microglial motility enabling interactions at neuron–astrocyte tripartite synapses. Abbreviations: STAT3 (signal transducer and activator of transcription 3), MERTK (MER tyrosine kinase), MEGF10 (multiple EGF-like domains 10), GLT1 (glutamate transporter 1), TGF-β (transforming growth factor beta), IL-33 (interleukin 33), C1q (complement Component 1q), NMDA (N-methyl-D-aspartate), AMPA (α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid), C3 (complement component 3), CR3 (complement receptor 3), and CR4 (complement receptor 4).
Figure 3. Diagram illustrating the mechanism of synaptic pruning in the CNS. Synaptic pruning is a key process in the developing and adult brain where excess or weak synapses are eliminated to refine neural circuits. In this mechanism, glutamate is released from the presynaptic terminal and binds to NMDA and AMPA receptors on the postsynaptic neuron, facilitating neurotransmission. Astrocytes maintain synaptic homeostasis by clearing excess glutamate via the GLT1 transporter. Meanwhile, signals like TGF-β from the presynaptic terminal induce the expression and relocalization of complement protein C1q, which, along with C3, tags synapses for elimination. These tags are recognized by CR3/CR4 receptors on microglial cells, which then engulf the marked synapses. Astrocytes also contribute to synaptic pruning through phagocytic receptors such as MERTK and MEGF10, regulated by the STAT3-SOCS3 pathway. Moreover, IL-33 released through astroglial end-feet further activates microglial pruning. The coordinated interplay of neurons, astrocytes, and microglial cells regulates synaptic remodeling during development, with microglial motility enabling interactions at neuron–astrocyte tripartite synapses. Abbreviations: STAT3 (signal transducer and activator of transcription 3), MERTK (MER tyrosine kinase), MEGF10 (multiple EGF-like domains 10), GLT1 (glutamate transporter 1), TGF-β (transforming growth factor beta), IL-33 (interleukin 33), C1q (complement Component 1q), NMDA (N-methyl-D-aspartate), AMPA (α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid), C3 (complement component 3), CR3 (complement receptor 3), and CR4 (complement receptor 4).
Biomedicines 14 00115 g003
Figure 4. Neuroinflammatory mechanisms underlying BBB disruption: glial cell activation, immune cell infiltration, and subsequent demyelination, with emphasis on potential therapeutic interventions targeting these pathological processes. Abbreviations: BBB (blood–brain barrier).
Figure 4. Neuroinflammatory mechanisms underlying BBB disruption: glial cell activation, immune cell infiltration, and subsequent demyelination, with emphasis on potential therapeutic interventions targeting these pathological processes. Abbreviations: BBB (blood–brain barrier).
Biomedicines 14 00115 g004
Table 3. Glia-targeted gene and RNA-based therapeutic strategies for modulating neurodegeneration and neuroinflammation. Abbreviations: AAV (adeno-associated virus), GFAP (glial fibrillary acidic protein), Iba1 (ionized calcium-binding adapter molecule 1 (Iba1), MBP (myelin basic protein), IGF-1 (insulin-like growth factor 1), BDNF (brain-derived neurotrophic factor), siRNA (small interfering RNA), ASO (antisense oligonucleotide), ALS (amyotrophic lateral sclerosis), BBB (blood–brain barrier), CNS (central nervous system), SOD1 (superoxide dismutase 1), and C9orf72 (chromosome 9 open reading frame 72).
Table 3. Glia-targeted gene and RNA-based therapeutic strategies for modulating neurodegeneration and neuroinflammation. Abbreviations: AAV (adeno-associated virus), GFAP (glial fibrillary acidic protein), Iba1 (ionized calcium-binding adapter molecule 1 (Iba1), MBP (myelin basic protein), IGF-1 (insulin-like growth factor 1), BDNF (brain-derived neurotrophic factor), siRNA (small interfering RNA), ASO (antisense oligonucleotide), ALS (amyotrophic lateral sclerosis), BBB (blood–brain barrier), CNS (central nervous system), SOD1 (superoxide dismutase 1), and C9orf72 (chromosome 9 open reading frame 72).
Therapeutic StrategyApproachTarget Glial CellsKey Effects/ApplicationsReferences
Viral vector-based gene deliveryAAV vectors with glial-specific promoters (GFAP, Iba1, and MBP)Astrocytes
Microglial cells
Oligodendrocytes
Cell-specific gene expression with long-term, and low-immunogenic
delivery
[305,306,307]
AAV-mediated IGF-1 or
BDNF overexpression
AstrocytesNeuroprotection, reduced
excitotoxicity, and synaptic support
[308,309]
Engineered AAVsMicroglial cellsShift toward anti-inflammatory and homeostatic phenotype[310]
RNA-based therapiessiRNAs and ASOsAstrocytes
Microglial cells
Neurons
Post-transcriptional gene silencing and transcript modulation[312,313]
ASOs targeting
Sod1 and C9orf72
Glial cells
Neurons
Reduction in toxic proteins
and RNA foci in ALS
[314,315]
Clinically optimized
ASOs/siRNAs
CNSBBB penetration and suitability
for repeated administration
[316,317]
Table 4. Glial cell transplantation strategies for neuroregeneration and restoration of CNS homeostasis. Abbreviations: iPSCs (induced pluripotent stem cells), OPCs (oligodendrocyte precursor cells), MLCs (microglia-like cells), ALSP (adult-onset leukoencephalopathy with axonal spheroids and pigmented glia), and GM2 (GM2 ganglioside).
Table 4. Glial cell transplantation strategies for neuroregeneration and restoration of CNS homeostasis. Abbreviations: iPSCs (induced pluripotent stem cells), OPCs (oligodendrocyte precursor cells), MLCs (microglia-like cells), ALSP (adult-onset leukoencephalopathy with axonal spheroids and pigmented glia), and GM2 (GM2 ganglioside).
Therapeutic
Strategy
ApproachTarget Glial CellsKey Effects/ApplicationsReferences
Astrocyte
transplantation
Astrocyte precursor cells
iPSC-derived astrocytes
AstrocytesImproved neuronal viability
Reduced gliosis
Axonal regeneration
Potential for personalized
autologous therapy
[318,319,320,321,322,323]
OPC
transplantation
Oligodendrocyte precursor cells
iPSC-derived OPCs
OligodendrocytesMigration to demyelinated regions
Differentiation and remyelination
Restoration of nerve conduction and functional improvement
[324,325,326,327,328]
Microglial
transplantation
Hematopoietic stem cells
iPSC-derived microglia
MLCs
Microglial cellsRestoration of CNS homeostasis
Myelin integrity
Reduction in axonal spheroids
Modulation of inflammation
Potential therapy for ALSP and Sandhoff disease
[329,330,331,332,333,334]
Table 5. Nanotechnology- and targeted delivery-based strategies for glia-focused modulation of neuroinflammation and CNS therapy. Abbreviations: BBB (blood–brain barrier), PNP (polymeric nanoparticle), RVG (rabies virus glycoprotein), siRNA (small interfering RNA), miRNA (microRNA), EV (extracellular vesicle), MSC (mesenchymal stem cell), EXOmotif (exosomal motif), TLR4 (Toll-like receptor 4), TOP1 (topoisomerase 1), and MS (multiple sclerosis).
Table 5. Nanotechnology- and targeted delivery-based strategies for glia-focused modulation of neuroinflammation and CNS therapy. Abbreviations: BBB (blood–brain barrier), PNP (polymeric nanoparticle), RVG (rabies virus glycoprotein), siRNA (small interfering RNA), miRNA (microRNA), EV (extracellular vesicle), MSC (mesenchymal stem cell), EXOmotif (exosomal motif), TLR4 (Toll-like receptor 4), TOP1 (topoisomerase 1), and MS (multiple sclerosis).
Therapeutic
Strategy
ApproachTarget Glial CellsKey Effects/ApplicationsReferences
Nanoparticle-mediated
delivery
PNPs
Liposomes
Astrocytes
Microglial cells
Cross BBB through receptor-
mediated transcytosis
Modulation of NF-κB, JAK/STAT, and MAPK pathways
[335,336,337,338,339,340,341,342,343,344,345]
Extracellular
vesicles/Exosomes
MSC-derived or
engineered exosomes
Astrocytes
Microglial cells
Modulation of neuroinflammation
Promotion of neuroprotection
Rewire glial phenotypes
[346,347,348,349,350,351,352]
BrainShuttle™ and nanobody platformsEngineered antibodies or nanobodies targeting BBB receptorsMicroglial cellsRgulation of pro-inflammatory
pathways
[353,354,355]
Targeted TOP1
inhibition
Topoisomerase 1 inhibitors (topotecan and
camptothecin) delivered through β-glucan-coated DNA origami (TopoGami)
Microglial cells
Myeloid cells
Suppression of microglial/
macrophage inflammation
Attenuation of neuroinflammation
[356]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

García-Domínguez, M. Glial Cell Dynamics in Neuroinflammation: Mechanisms, Interactions, and Therapeutic Implications. Biomedicines 2026, 14, 115. https://doi.org/10.3390/biomedicines14010115

AMA Style

García-Domínguez M. Glial Cell Dynamics in Neuroinflammation: Mechanisms, Interactions, and Therapeutic Implications. Biomedicines. 2026; 14(1):115. https://doi.org/10.3390/biomedicines14010115

Chicago/Turabian Style

García-Domínguez, Mario. 2026. "Glial Cell Dynamics in Neuroinflammation: Mechanisms, Interactions, and Therapeutic Implications" Biomedicines 14, no. 1: 115. https://doi.org/10.3390/biomedicines14010115

APA Style

García-Domínguez, M. (2026). Glial Cell Dynamics in Neuroinflammation: Mechanisms, Interactions, and Therapeutic Implications. Biomedicines, 14(1), 115. https://doi.org/10.3390/biomedicines14010115

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop