Fact-checked by Grok 2 weeks ago

Gliosis

Gliosis is a fundamental reactive process in the central nervous system (CNS) characterized by the activation, proliferation, and hypertrophy of glial cells—primarily astrocytes and microglia—in response to injury, infection, or disease, serving as a nonspecific defense mechanism to maintain tissue integrity and limit damage spread. This response, commonly termed reactive gliosis, involves morphological changes such as upregulation of glial fibrillary acidic protein (GFAP) in astrocytes and ionized calcium-binding adapter molecule 1 (Iba1) in microglia, enabling the formation of a glial scar that physically barriers the lesion site while modulating local inflammation and immune responses.
In the acute phase following CNS insult, gliosis promotes neuroprotection by isolating necrotic tissue, scavenging debris, and supporting neuronal survival through the release of neurotrophic factors, though excessive or prolonged activation can transition to a detrimental state by inhibiting axonal regrowth and contributing to chronic neurodegeneration.
Reactive gliosis manifests in heterogeneous forms tailored to the insult's nature and severity, including astrogliosis, where astrocytes can adopt various reactive phenotypes, including the proposed neurotoxic A1 subtype that promotes inflammation and neuronal death and the neuroprotective A2 subtype that enhances synaptic support, though these represent a simplification of a more continuous spectrum—and microgliosis, involving microglial activation encompassing a spectrum of states, often described in simplified terms as pro-inflammatory M1 or anti-inflammatory M2 polarization.
These glial transformations are triggered by diverse pathologies, including traumatic brain injury, stroke, neurodegenerative disorders like Alzheimer's disease, and infections, with the process being highly context-dependent and influenced by factors such as cytokine signaling from activated microglia.
Oligodendrocytes and NG2-glia may also participate, contributing to remyelination efforts or scar formation, underscoring gliosis as a multicellular, dynamic response rather than a uniform event.
The dual-edged nature of gliosis—beneficial for acute repair yet potentially pathological in chronic contexts—has positioned it as a key target for therapeutic interventions, with research exploring modulation via genetic, pharmacological, or molecular approaches to harness its protective aspects while mitigating inhibitory effects on CNS recovery.
Understanding gliosis's spatiotemporal progression across brain regions, such as the hippocampus or cortex, remains crucial for addressing its role in long-term outcomes like cognitive impairment following insults.

Overview and Fundamentals

Definition and Historical Context

Gliosis is a reactive process in the central nervous system (CNS) characterized by the proliferation, hypertrophy, and functional alterations of glial cells in response to injury, infection, or degenerative conditions, often culminating in the formation of a glial scar that isolates damaged tissue. This response involves both morphological changes, such as cellular enlargement and process extension, and molecular shifts that enable glia to support repair and limit further damage. Unlike direct neuronal cell death mechanisms, gliosis represents a non-lethal, adaptive reaction aimed at maintaining CNS homeostasis. The concept of gliosis traces its origins to 19th-century neuropathology, where Rudolf Virchow first described neuroglia in 1846 as a connective tissue-like substance in post-mortem brains, likening it to a "nerve glue" that formed scar-like structures following injury. Early understandings framed gliosis primarily as a static pathological endpoint observed in autopsies, reflecting the limitations of histological techniques at the time. A pivotal advancement came in 1919 with Pío del Río-Hortega's classification of microglia as a distinct glial lineage using silver staining methods, expanding recognition beyond astrocytes to include immune-active glia in reactive processes. By the 1980s, research shifted toward viewing reactive gliosis as a dynamic, multifaceted phenomenon with both protective and inhibitory roles, influenced by advances in immunohistochemistry and in vivo imaging that revealed ongoing cellular interactions. This evolution highlighted gliosis not merely as scar formation but as an orchestrated response involving astrocytes, microglia, and other glia, as explored in subsequent sections on specific cell types. In contrast to necrosis, which involves uncontrolled cell swelling and rupture leading to inflammation, or apoptosis, a programmed neuronal demise, gliosis preserves glial viability to facilitate long-term tissue remodeling.

Types of Glial Cells Involved

Astrocytes are the most abundant glial cells in the central nervous system (CNS) and play essential roles in maintaining the blood-brain barrier (BBB) by inducing endothelial tight junctions and regulating nutrient transport across it. They also provide structural and metabolic support to synapses, facilitating synapse formation, maturation, and pruning through the release of trophic factors and uptake of excess neurotransmitters like glutamate. Additionally, astrocytes contribute to neurotransmitter homeostasis by buffering ions such as potassium and modulating synaptic transmission via gliotransmitter release. In the context of gliosis, astrocytes predominate among the reactive glial population, undergoing characteristic changes that form a major component of the glial scar. Microglia function as the resident immune cells of the CNS, originating from yolk sac progenitors and maintaining a ramified morphology for constant surveillance of the neural parenchyma to detect pathogens, debris, or neuronal distress. They perform phagocytosis to eliminate apoptotic cells, synaptic remnants, and aggregated proteins, thereby preserving tissue homeostasis and supporting neuronal circuit refinement during development and adulthood. In gliosis, microglia rapidly activate, transitioning to an amoeboid shape and proliferating to orchestrate the early inflammatory aspects of the response alongside astrocytes. Oligodendrocytes are specialized glial cells responsible for producing the myelin sheath that insulates CNS axons, enabling saltatory conduction for efficient neural signaling, and providing metabolic support by supplying lactate and other substrates to axons under high demand. While their primary role is myelination, oligodendrocyte precursor cells (OPCs) can proliferate and contribute to reactive gliosis, though their overall involvement remains secondary and less pronounced compared to astrocytes and microglia. Other glial cell types, such as ependymal cells, line the brain ventricles and central canal, where they facilitate cerebrospinal fluid (CSF) production and circulation while forming a barrier to protect the CNS from pathogens in the CSF. These cells may exhibit limited reactive changes akin to astrogliosis in response to injury, but their contribution to gliosis is minor. In contrast, Schwann cells, which myelinate peripheral nervous system (PNS) axons and promote regeneration there, do not participate in CNS gliosis, underscoring the distinct glial responses between the CNS and PNS. Astrocytes and microglia constitute the primary actors in the gliosis response, comprising the majority of reactive changes in the CNS, with oligodendrocytes and other glia playing supportive or secondary roles.

Astrogliosis

Cellular and Morphological Changes

During astrogliosis, astrocytes undergo prominent morphological transformations characterized by hypertrophy, where the cell body enlarges and cellular processes thicken substantially. These changes are often visible within days following an insult, such as after an entorhinal cortex lesion, and help maintain the astrocytes' tiled territorial domains with minimal overlap between neighboring cells. A hallmark of this reactivity is the robust upregulation of glial fibrillary acidic protein (GFAP), an intermediate filament that reinforces the cytoskeletal structure and serves as a primary marker for identifying reactive astrocytes. In addition to hypertrophy, reactive astrocytes exhibit increased expression of other intermediate filaments, including vimentin and nestin, which further stabilize the altered morphology and support process extension. Functional shifts accompany these structural changes, such as the loss of gap junction coupling mediated by reduced connexin 43 expression, which disrupts the astrocytic syncytium and alters intercellular communication. Reactive astrocytes also enhance production of extracellular matrix components, notably chondroitin sulfate proteoglycans, contributing to the formation of a dense glial scar through proliferation of a subset of these cells, particularly those near lesion sites or blood vessels. The progression of these cellular changes follows a temporal sequence, with an acute phase occurring within hours to days post-insult, marked by upregulation of immediate early genes like c-fos that initiate transcriptional responses in astrocytes. This is followed by a chronic phase spanning weeks, during which the glial scar stabilizes and the morphological alterations become more persistent. Astrogliosis displays regional heterogeneity in these transformations, with greater proliferative responses observed in white matter astrocytes compared to those in gray matter, reflecting differences in their baseline fibrous versus protoplasmic morphologies.

Molecular Mechanisms of Modulation

The molecular mechanisms modulating astrogliosis involve intricate signaling pathways that control astrocyte proliferation, hypertrophy, and inflammatory responses following central nervous system (CNS) injury. These pathways are activated by extracellular signals and integrate to orchestrate reactive changes in astrocytes, such as upregulation of glial fibrillary acidic protein (GFAP). Key signaling cascades include the Janus kinase-signal transducer and activator of transcription 3 (JAK-STAT3) pathway, which drives astrocyte proliferation and hypertrophy. Cytokines like interleukin-6 (IL-6) bind to glycoprotein 130 (gp130) receptors, activating JAK kinases that phosphorylate STAT3, leading to its nuclear translocation and transcription of genes promoting cell division and GFAP expression. Similarly, the transforming growth factor-β (TGF-β)/Smad pathway regulates glial scar formation by inducing extracellular matrix components; TGF-β ligands activate Smad proteins, which dimerize and translocate to the nucleus to upregulate scar-associated genes like chondroitin sulfate proteoglycans. The nuclear factor-κB (NF-κB) pathway contributes to inflammatory modulation, where injury signals trigger IκB kinase-mediated release of NF-κB dimers, enabling their nuclear entry and expression of pro-inflammatory cytokines in reactive astrocytes. Positive modulators of astrogliosis encompass cytokines such as IL-6 and interleukin-1β (IL-1β), which amplify JAK-STAT3 and NF-κB signaling to enhance astrocyte reactivity. Growth factors including ciliary neurotrophic factor (CNTF) and leukemia inhibitory factor (LIF) also activate gp130-coupled receptors, converging on JAK-STAT3 to promote proliferation and morphological changes. Damage-associated molecular patterns (DAMPs) like high-mobility group box 1 (HMGB1), released from damaged cells, act as alarmin signals that stimulate Toll-like receptor 4 (TLR4) on astrocytes, initiating NF-κB-dependent inflammatory responses and astrocyte activation. Negative regulators counteract excessive astrogliosis to maintain balance. Suppression of phosphatase and tensin homolog (PTEN), a key inhibitor of the PI3K/Akt/mTOR pathway, enhances astrocyte hypertrophy and proliferation, as demonstrated in conditional PTEN knockout models where astrocytes exhibit increased size and mitotic activity. MicroRNAs such as miR-29 inhibit astrogliosis by targeting pro-apoptotic and scar-promoting genes; miR-29a knockout exacerbates astrocyte proliferation and glutamate release post-ischemia, while its overexpression protects against reactive changes. Pharmacologically, minocycline attenuates astrogliosis by inhibiting microglial-derived signals that activate astrocytic NF-κB and matrix metalloproteinases (MMPs), reducing GFAP expression and scar formation in injury models. Feedback loops sustain astrogliosis through autocrine mechanisms, notably endothelin-1 (ET-1), which is upregulated in reactive astrocytes and binds ET_B receptors to activate JNK/c-Jun signaling, promoting further proliferation and GFAP induction in a self-amplifying manner.

Beneficial Functions

Reactive astrocytes contribute to neuroprotection during astrogliosis by forming a glial scar that physically isolates the lesion site, thereby containing inflammatory cells, excitotoxic molecules, and other neurotoxic factors to prevent their diffusion into surrounding healthy tissue. This barrier function is evident in models of focal cerebral ischemia and traumatic brain injury, where the scar limits the expansion of secondary damage. Furthermore, reactive astrocytes upregulate antioxidant enzymes, such as superoxide dismutase (SOD) and glutathione peroxidase (GPx), to counteract oxidative stress generated by reactive oxygen species in the injured environment. These protective effects are associated with the adoption of a neuroprotective A2 astrocyte phenotype, characterized by upregulation of factors like S100A10, thrombospondins, and Clq, which promote synaptogenesis, debris clearance, and neuronal survival. In contrast, the neurotoxic A1 phenotype, marked by expression of C3 and other complement components, drives inflammation and neuronal death; however, in acute beneficial contexts, A2 polarization predominates to support repair. This binary classification, while useful, oversimplifies astrocyte heterogeneity, as recent studies (as of 2024) emphasize context-dependent and regional variations in reactive states. Astrogliosis also supports tissue repair through the secretion of neurotrophic factors, including brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF), which enhance neuronal survival, promote synaptogenesis, and facilitate axonal remodeling in the peri-lesion area. In addition, these cells release vascular endothelial growth factor (VEGF) to aid vascular remodeling, including angiogenesis and blood-brain barrier repair, which are critical for restoring cerebral perfusion and nutrient supply post-injury. Experimental evidence highlights the beneficial impact of astrogliosis; for instance, GFAP knockout mice exhibit larger infarct volumes and impaired neurological recovery in middle cerebral artery occlusion stroke models compared to wild-type controls, indicating that attenuated astrocytic reactivity exacerbates damage. In early phases of traumatic brain injury, reactive astrogliosis similarly promotes recovery by suppressing excessive inflammation and supporting neurogenesis via factors like S100β. Quantitative assessments in rodent ischemia studies show that the glial scar limits lesion expansion, as demonstrated by increased lesion sizes in astrocyte-compromised models.

Detrimental Consequences

While reactive astrogliosis initially limits lesion expansion, it can become maladaptive by forming a glial scar that serves as a physical and chemical barrier to axonal regeneration. Reactive astrocytes within the scar upregulate the production of chondroitin sulfate proteoglycans (CSPGs), extracellular matrix molecules that inhibit axonal sprouting and neurite outgrowth by interacting with growth cone receptors and disrupting cytoskeletal dynamics. This inhibitory environment persists in the chronic phase, preventing regenerative efforts in conditions like spinal cord injury where CSPG levels remain elevated for months post-insult. A key detrimental outcome of prolonged astrogliosis is the promotion of chronic inflammation through sustained cytokine release, particularly tumor necrosis factor-α (TNF-α), which exacerbates neuronal damage via excitotoxicity. Reactive astrocytes secrete TNF-α in response to injury signals, leading to reduced glutamate uptake by downregulating excitatory amino acid transporter 2 (EAAT2) expression and increasing extracellular glutamate accumulation. This, in turn, heightens calcium influx through AMPA and NMDA receptors on neurons, contributing to secondary neurodegeneration in both acute injuries and chronic pathologies such as multiple sclerosis. These neurotoxic effects are linked to the A1 astrocyte phenotype, which upregulates pro-inflammatory genes like C3 and may predominate in chronic or severe insults, though astrocyte responses are highly heterogeneous and not strictly binary. Experimental evidence from genetic ablation models underscores the dual-edged nature of astrogliosis, where suppressing reactive astrocytes worsens immediate tissue damage but can facilitate improved long-term neural plasticity. In spinal cord injury paradigms using conditional knockout of signal transducer and activator of transcription 3 (STAT3) or suppressor of cytokine signaling 3 (SOCS3) in astrocytes, early impairment of astrogliosis results in unchecked inflammation, larger cystic cavities, and heightened immune infiltration acutely. However, this intervention reduces scar-mediated inhibition over time, promoting greater axonal sprouting and functional recovery in chronic phases compared to unablated controls. The transition from acute protective responses to chronic fibrotic pathology in astrogliosis involves epigenetic reprogramming, notably alterations in histone acetylation that lock in the reactive phenotype. Increased global histone H4 acetylation in reactive astrocytes correlates with persistent upregulation of glial fibrillary acidic protein (GFAP) and pro-fibrotic genes, sustaining scar formation and inhibiting resolution. Sustained STAT3 activation further exacerbates this progression by driving epigenetic modifiers toward a profibrotic state.

Microgliosis

Activation States and Morphological Features

Microglia in their resting state, often denoted as M0, exhibit a ramified morphology characterized by elongated processes with fine branching that facilitate constant surveillance of the neural parenchyma. Upon activation during gliosis, these cells transition through a spectrum of states rather than a strict binary classification, with classical M1-like pro-inflammatory activation and alternative M2-like anti-inflammatory activation representing extremes of a continuum influenced by environmental cues. Recent studies emphasize the heterogeneity of these states, including context-specific profiles such as disease-associated microglia (DAM) in neurodegenerative conditions, which exhibit unique transcriptional signatures combining pro- and anti-inflammatory functions. The M1 state is associated with heightened production of pro-inflammatory cytokines such as TNF-α and IL-1β, promoting phagocytosis of debris but potentially exacerbating tissue damage, while the M2 state involves anti-inflammatory factors like IL-10 and TGF-β to support resolution and repair. Morphological features undergo distinct changes during activation, including retraction of processes, enlargement of the cell soma, and adoption of an amoeboid shape that enhances motility and phagocytic capacity. These transformations are accompanied by upregulation of ionized calcium-binding adapter molecule 1 (Iba1), a reliable marker for microglial activation detectable via immunohistochemistry, which reflects cytoskeletal remodeling and increased protein expression. In the M1 phenotype, the amoeboid form is more pronounced with rounded somata and short, thick processes, whereas M2 activation may retain some branching or display a bushy appearance to aid in tissue remodeling. Activation initiates rapidly, often within minutes to hours following injury, triggered by purinergic signaling from ATP release by damaged cells, which binds to P2Y and P2X receptors on microglia to initiate migration and proliferation. This acute response can resolve in healthy contexts, returning microglia toward a surveillant state, or persist as chronic priming in pathological conditions, leading to sustained morphological alterations. Microglial activation displays significant heterogeneity, with disease-specific profiles; for instance, in Alzheimer's disease, there is a predominance of M1-like states characterized by enhanced pro-inflammatory signaling and amyloid plaque association, contrasting with more balanced M2 responses in other contexts.

Neural and Immune Regulation

Microglial activation during gliosis is tightly regulated by neural signals that maintain a balance between protective surveillance and potentially harmful inflammation. Neuronal fractalkine (CX3CL1), expressed on neuronal membranes, binds to the CX3CR1 receptor on microglia, inhibiting excessive activation and promoting a ramified morphology to preserve tissue homeostasis. In contrast, purinergic signaling through the P2X7 receptor, activated by extracellular ATP released from damaged neurons, drives a pro-inflammatory shift in microglia, enhancing cytokine release and contributing to reactive states. Immune inputs further modulate microglial responses in gliosis via pattern recognition receptors and cytokines. Toll-like receptor 4 (TLR4) on microglia detects lipopolysaccharide (LPS) from pathogens or damage-associated molecular patterns (DAMPs) from injured cells, triggering NF-κB signaling and pro-inflammatory activation. Cytokine priming by interferon-γ (IFN-γ), often from infiltrating T cells, sensitizes microglia toward an M1-like phenotype, amplifying responses to secondary stimuli and sustaining inflammation. Regulatory mechanisms ensure balanced microglial function, with receptors like TREM2 promoting phagocytosis of debris without excessive inflammation, thus supporting debris clearance in gliotic environments. The CD200-CD200R interaction, where neuronal CD200 ligand engages microglial CD200R, suppresses activation by inhibiting pro-inflammatory pathways such as NF-κB, fostering an anti-inflammatory state. Recent studies highlight the CX3CL1/CX3CR1 axis's role in modulating gliosis severity during neurodegeneration, where its signaling dampens microglial overactivation and reduces associated neuronal loss.

Crosstalk with Astrogliosis

The bidirectional crosstalk between microglia and astrocytes is a critical aspect of gliosis, where these cells exchange signals to either amplify reactive responses or promote resolution following central nervous system (CNS) injury. Activated microglia release pro-inflammatory cytokines such as interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α), which directly induce astrocyte reactivity by upregulating glial fibrillary acidic protein (GFAP) expression and promoting the transition to a pro-inflammatory A1 astrocyte phenotype. This signaling pathway enhances astrogliosis, contributing to the containment of damage but potentially exacerbating neuroinflammation if unchecked. Additionally, microglial phagocytosis plays a regulatory role by efficiently clearing cellular debris and apoptotic cells, thereby reducing persistent danger signals that could otherwise sustain prolonged astrocyte activation and limit the extent of reactive gliosis. In the reverse direction, astrocytes modulate microglial states through secreted factors that favor anti-inflammatory phenotypes. Astrocyte-derived transforming growth factor-β (TGF-β) promotes a shift in microglia toward the M2 (anti-inflammatory) polarization, suppressing pro-inflammatory cytokine production and enhancing tissue repair processes during the subacute phase of injury. Furthermore, astrocytes release ATP, which activates P2Y12 receptors on microglia, facilitating their process extension, migration to injury sites, and targeted phagocytosis without necessarily driving excessive inflammation. This interaction underscores the astrocytes' role in fine-tuning microglial responses to prevent overactivation. The synergistic effects of co-activated microglia and astrocytes often culminate in glial scar formation, where microglial cytokines trigger astrocyte proliferation and extracellular matrix deposition, forming a protective barrier that isolates the lesion core. Disruptions in this crosstalk, such as loss of TREM2 in microglia, impair phagocytic efficiency and cytokine regulation, leading to exacerbated reactivity in both cell types, increased neuroinflammation, and worsened outcomes in models of neurodegeneration and injury. In vitro co-culture studies of microglia and astrocytes demonstrate this interplay, revealing increased production of pro-inflammatory cytokines like IL-6 and TNF-α upon combined activation compared to monocultures, highlighting the amplifying potential of their interactions.

Oligodendroglial and Other Glial Responses

Reactive Changes in Oligodendrocytes

Oligodendrocytes, the myelinating cells of the central nervous system, display reactive changes following injury that are less pronounced than those in astrocytes or microglia but play a critical role in attempted repair processes. These changes primarily involve oligodendrocyte progenitor cells (OPCs), which proliferate and migrate to lesion sites to facilitate remyelination, though success is often limited by the injury environment. Unlike the scar-forming responses of other glia, oligodendroglial reactivity focuses on myelin restoration. A key feature of these reactive changes is the upregulation of NG2, a chondroitin sulfate proteoglycan, in OPCs, which promotes their proliferation and migration toward injury sites. Following spinal cord injury or demyelination, NG2 expression increases in both OPCs and reactive macrophages, enabling OPCs to expand their population and contribute to tissue remodeling. This upregulation is part of a multicellular response, where NG2-positive OPCs actively invade lesion areas, though excessive NG2 may also contribute to inhibitory extracellular matrix formation. Mature oligodendrocytes undergo sublethal injury characterized by process retraction and downregulation of myelin-related genes such as myelin basic protein (MBP), driven by inflammatory signals like IFN-γ and TNF-α. This downregulation of MBP and other myelin genes, observed in sublethal injury models, reflects metabolic stress and integrated stress response activation. Functionally, reactive oligodendrocytes shift toward attempted remyelination, with OPCs differentiating to replace lost myelin sheaths, though this process is hindered in chronic conditions. In chronic gliosis, oligodendrocytes exhibit increased susceptibility to apoptosis, exacerbated by neuroinflammatory cytokines (e.g., TNF-α, IL-1β) and oxidative stress, leading to progressive white matter loss. Olig2, a transcription factor marker of the oligodendrocyte lineage, persists in surviving cells amid injury, maintaining lineage identity even as cells undergo stress. However, reactive gliosis from astrocytes inhibits OPC maturation through chondroitin sulfate proteoglycans (CSPGs), such as versican, which reduce OPC process outgrowth and differentiation in a dose-dependent manner. Astrocytic and microglial barriers, including CSPG deposition, further limit remyelination by blocking OPC access to axons. In multiple sclerosis (MS) models, this gliosis-associated inhibition correlates with remyelination failure, where OPC differentiation blocks result in substantially reduced efficiency, often below 50% in chronic lesions.

Responses in Ependymal and Other Glia

Ependymal cells, which form a ciliated epithelial lining of the brain's ventricular system, undergo significant reactive changes during gliosis, particularly following traumatic brain injury (TBI). A hallmark of this response is the rapid and persistent loss of motile cilia on their apical surfaces, which disrupts cerebrospinal fluid (CSF) flow and contributes to ventricular dilation. This ciliary dysfunction is accompanied by denudation of the ependymal layer, where cells lose their tight junctions and epithelial integrity, leading to barrier breaches that exacerbate neuroinflammation. In response, surviving ependymal cells proliferate extensively to repair these breaches, migrating to reseal the lining and restore barrier function, a process critical for limiting secondary damage in the periventricular region. Morphological alterations in reactive ependymal cells include a shift toward astrocyte-like features, such as upregulation of glial fibrillary acidic protein (GFAP), mirroring aspects of astrogliosis while maintaining their role in CSF homeostasis. Recent studies in hydrocephalus models have highlighted ependymal gliosis as a key pathological feature, with 2024 research demonstrating glial remodeling involving astrogliosis and persistent neuroinflammation triggered by inflammatory cues, which may worsen ventricular enlargement but also support tissue repair. For instance, in rodent models of congenital hydrocephalus, ependymal denudation triggers periventricular gliosis, where reactive ependyma contribute to scar formation around the ventricles. Beyond ependyma, other specialized glia exhibit gliotic responses tailored to their niches. In the cerebellum, Bergmann glia—radial astrocytes intimately associated with Purkinje neurons—undergo reactive gliosis characterized by GFAP upregulation and process hypertrophy, enhancing structural support for Purkinje cell survival and synaptic stability during injury or degeneration. This response helps maintain cerebellar circuitry but can lead to excessive scarring if prolonged. In contrast, peripheral nervous system (PNS) Schwann cells show more robust regenerative potential analogous to gliosis, rapidly dedifferentiating and proliferating to clear debris and remyelinate axons, highlighting regional differences in glial reactivity between CNS and PNS. Functionally, reactive ependymal cells contribute to gliosis by preserving CSF circulation through residual ciliary beating and barrier reformation, preventing hydrocephalus progression. Additionally, ependymal cells in the subventricular zone (SVZ) harbor stem-like properties, activating upon injury to generate neural progenitors that aid in limited tissue repair and neurogenesis. These roles underscore their supportive position in the broader gliotic response, bridging epithelial integrity with neurogenic potential.

Triggers of Gliosis

Acute Injury Triggers

Acute injury triggers of gliosis primarily arise from sudden disruptions to the central nervous system (CNS), such as physical trauma, oxygen deprivation, or pathogen invasion, which rapidly activate glial cells through the release of signaling molecules. Mechanical trauma, often involving shear forces during events like traumatic brain injury (TBI), initiates gliosis by disrupting cellular membranes and releasing adenosine triphosphate (ATP) and ion fluxes from damaged neurons and glia. This ATP release acts as a danger signal, binding to purinergic receptors on astrocytes and microglia to promote their activation and proliferation. Additionally, mechanical injury compromises the blood-brain barrier (BBB), allowing influx of blood components and further inflammatory mediators that exacerbate glial responses. Ischemia and hypoxia, as seen in stroke, trigger gliosis through metabolic stress, including lactate accumulation from anaerobic glycolysis and reactive oxygen species (ROS) generated by mitochondrial dysfunction. Elevated lactate levels directly promote astrocyte reactivity, enhancing glycolytic pathways and contributing to cellular hypertrophy. Meanwhile, mitochondrial impairment during oxygen deprivation leads to excessive ROS production, which damages cellular components and signals glial activation via oxidative stress pathways. Infections serve as acute triggers when pathogen-associated molecular patterns (PAMPs), such as viral double-stranded RNA (dsRNA), are recognized by glial cells. For instance, dsRNA from viral infections activates retinoic acid-inducible gene I (RIG-I) in astrocytes, initiating type I interferon signaling and subsequent gliosis to contain the pathogen. A common pathway underlying these acute triggers involves damage-associated molecular patterns (DAMPs), such as S100B released from injured cells, which bind to Toll-like receptors (TLRs) on glia to propagate inflammatory cascades and gliotic responses. This DAMP-mediated TLR activation integrates signals from diverse injuries, unifying the initiation of reactive gliosis across mechanical, ischemic, and infectious insults.

Chronic and Pathological Triggers

Chronic gliosis in the central nervous system can be sustained by persistent protein aggregates, such as amyloid-beta (Aβ) and tau in Alzheimer's disease (AD), which activate receptors like the receptor for advanced glycation end products (RAGE) on glial cells, leading to prolonged inflammatory signaling and reactive astrogliosis. Aβ oligomers bind to RAGE, promoting microglial activation and the release of pro-inflammatory cytokines that exacerbate gliosis around plaques. Similarly, hyperphosphorylated tau aggregates induce astrocytic and microglial responses, contributing to a self-perpetuating cycle of neuroinflammation and glial proliferation in tauopathies. In Parkinson's disease (PD), α-synuclein aggregates trigger microglial activation via Toll-like receptors, fostering a chronic neuroinflammatory environment that sustains gliosis in the substantia nigra. Metabolic disturbances, particularly those induced by high-fat diets, drive hypothalamic gliosis linked to obesity through mechanisms involving leptin resistance. Prolonged exposure to high-fat diets elevates circulating leptin levels, but chronic hypothalamic inflammation impairs leptin receptor signaling, resulting in sustained astrogliosis and microgliosis that disrupt energy homeostasis. This gliotic response, characterized by increased glial fibrillary acidic protein expression, correlates with insulin resistance and perpetuates metabolic dysfunction in the mediobasal hypothalamus. Recent reviews highlight how this process forms a feedback loop, where gliosis further desensitizes leptin-sensitive neurons, maintaining the obese state. In autoimmune conditions like multiple sclerosis (MS), exposure to myelin antigens, such as myelin basic protein, initiates complement activation that amplifies gliosis. Autoantibodies against myelin components bind to oligodendrocytes and axons, recruiting complement proteins like C3 and C5b-9, which deposit in plaques and trigger reactive gliosis in surrounding astrocytes and microglia. This complement-mediated inflammation sustains chronic demyelination and glial activation, contributing to the progressive nature of MS pathology. The persistence of gliosis in these pathological states is reinforced by epigenetic modifications, including DNA methylation, which lock in pro-inflammatory glial phenotypes. Aberrant hypermethylation of promoter regions in glial cells suppresses anti-inflammatory genes while enhancing expression of cytokines and glial activation markers, thereby maintaining the chronic reactive state. In models of neurodegenerative disease, such epigenetic changes in astrocytes and microglia prevent resolution of gliosis, promoting long-term neuroinflammation.

Gliosis in CNS Injury and Disease

Traumatic Brain and Spinal Cord Injury

Traumatic brain injury (TBI) induces a rapid response, particularly in the cortical regions surrounding the lesion. Microgliosis in the cortex typically peaks within 1-3 days post-injury, driven by the of resident to clear through and initiate inflammatory signaling. Astrogliosis follows, with reactive forming a dense at the lesion core that compartmentalizes damage but can impede tissue remodeling depending on injury severity. The extent of this gliosis varies with TBI severity, as milder impacts lead to transient , while severe contusions result in prolonged scarring that influences long-term neurodegeneration. In spinal cord injury (SCI), gliosis exhibits a rostro-caudal gradient, with the most intense reactive changes concentrated at the injury epicenter and diminishing toward rostral and caudal regions. This pattern arises from the focal nature of mechanical trauma, where microglia and astrocytes proliferate asymmetrically to isolate necrotic tissue. The resulting inhibitory glial scar at the epicenter, rich in chondroitin sulfate proteoglycans, physically and chemically hinders axon regrowth by creating a non-permissive environment for sprouting. Unlike in TBI, SCI gliosis often persists more robustly along the cord's axis, contributing to functional deficits below the injury site. Gliosis after traumatic CNS injury unfolds in distinct stages: an acute phase dominated by microglial phagocytosis of cellular debris within hours to days, a subacute repair phase involving astrocytic proliferation and scar maturation from 2-14 days, and a chronic fibrosis stage marked by persistent extracellular matrix deposition beyond weeks. These stages reflect a shift from protective inflammation to barrier formation, though excessive fibrosis can exacerbate axon inhibition. In humans, gliosis following TBI and SCI can be detected non-invasively through elevated glial fibrillary acidic protein (GFAP) levels in serum or cerebrospinal fluid, which correlate with MRI findings such as lesion volume and white matter disruption. Higher GFAP concentrations predict poorer neurological outcomes, including motor impairment and cognitive deficits, providing a biomarker for injury severity and recovery prognosis in both TBI and SCI cohorts.

Neuroinflammatory and Autoimmune Disorders

In multiple sclerosis (MS), a prototypical neuroinflammatory and autoimmune disorder, gliosis manifests prominently around demyelinating plaques, where perivascular microgliosis and astrocytic proliferation contribute to lesion pathology. Reactive microglia form clusters or nodules adjacent to active plaques, often in perivascular locations, exacerbating local inflammation through phagocytosis of myelin debris and cytokine release. Concurrently, astrocytes undergo hypertrophy and proliferation, forming a dense glial scar that encapsulates the lesion and limits axonal regeneration. These astrocytic changes, characterized by upregulation of intermediate filaments like GFAP, create a physical and biochemical barrier that inhibits remyelination by blocking oligodendrocyte progenitor cell migration and differentiation. Experimental autoimmune encephalomyelitis (EAE), an animal model of MS, further illustrates immune-driven gliosis mechanisms, particularly through complement component C3 produced by myeloid cells. In a 2025 study, myeloid-derived C3 was shown to induce reactive gliosis during the early inflammatory phase of EAE, promoting disease-associated glial subtypes with heightened expression of pro-inflammatory genes and neuronal stress markers. Deletion of C3 mitigated these glial alterations, restoring more homeostatic profiles and reducing optic nerve pathology, highlighting C3's role in amplifying gliotic responses in autoimmune contexts. Gliosis in these disorders exhibits distinct morphological features, ranging from focal clusters of activated microglia and astrocytes around acute lesions to diffuse, fibrotic scars in chronic phases. Interferon-gamma (IFN-γ) plays a key amplifying role, enhancing microglial reactivity and astrocytic signaling to sustain inflammation, as evidenced in EAE where regional IFN-γ in astrocytes promotes protective yet prolonged gliotic responses. Oligodendrocytes in plaques may show reactive changes, but these are secondary to the dominant astro- and microgliosis. The progression of gliosis mirrors the clinical from relapsing-remitting to active states, with acute responses evolving into persistent scarring that drives neurodegeneration. In relapsing-remitting , episodic triggers transient gliosis that partially resolves, but over time, unresolved leads to active lesions with ongoing microglial rims and astrocytic borders, correlating with accumulation. This transition underscores gliosis as a maladaptive in autoimmune neuroinflammation, where early immune dysregulation perpetuates glial hyperactivity.

Neurodegenerative Diseases

In neurodegenerative diseases characterized by proteinopathies, gliosis emerges as a secondary reactive process that amplifies neuronal damage through neuroinflammation and impaired homeostasis. Microglial activation and astrogliosis often correlate with the accumulation of misfolded proteins, such as amyloid-beta (Aβ) in Alzheimer's disease (AD) and TDP-43 in amyotrophic lateral sclerosis (ALS), contributing to progressive degeneration without directly initiating the pathology. These glial responses, while initially protective via phagocytosis and cytokine signaling, shift toward neurotoxicity in chronic stages, exacerbating synaptic loss and circuit dysfunction across disorders like Parkinson's disease (PD) and Huntington's disease (HD). In AD, Aβ plaques induce pronounced microgliosis, with activated microglia proliferating and clustering around plaques to facilitate phagocytosis but also releasing proinflammatory cytokines that propagate pathology. This Aβ-driven microglial response creates a microenvironment conducive to tau seeding in dystrophic neurites, enhancing neurofibrillary tangle formation. Concurrently, astrogliosis correlates strongly with tau spread, as reactive astrocytes upregulate tau kinase-activating factors like TGF-β1 and exhibit impaired clearance, accelerating hyperphosphorylation and propagation from entorhinal cortex to hippocampus. Senescent astrocytes further worsen tau-mediated neurodegeneration by promoting neurofibrillary tangle burden, underscoring gliosis as a modulator of AD progression. In ALS, microgliosis is intimately linked to motor neuron vulnerability, particularly in SOD1 mutation carriers, where mutant SOD1 expression in microglia drives non-cell-autonomous toxicity via excessive TNF-α and NO production, hastening denervation and paralysis in preclinical models. TDP-43 aggregates, pathological hallmarks in over 97% of sporadic ALS cases, trigger microglial phagocytosis through TREM2-dependent pathways, but dysfunctional clearance leads to persistent inflammation and motor neuron loss in the spinal cord and cortex. This motor neuron-centric gliosis correlates with upper motor neuron symptoms, as evidenced by PET imaging showing early microglial activation in presymptomatic stages. In PD, dopaminergic gliosis manifests in the substantia nigra, where α-synuclein aggregates activate microglia via TLR2/NF-κB signaling prior to significant neuron loss, fostering a proinflammatory milieu that impairs dopamine homeostasis. Astrocytes contribute by accumulating pathologic α-synuclein, disrupting glutamate uptake and releasing IL-1β/IL-6, which amplifies dopaminergic degeneration. In HD, striatal astro-microglial interactions drive pathology, with mutant huntingtin in microglia upregulating PU.1 transcription factors to enhance proinflammatory profiles, while activated microglia induce A1-like neurotoxic astrocytes that downregulate EAAT2 transporters, leading to excitotoxicity and medium spiny neuron loss. These bidirectional glial communications in the striatum correlate with disease severity, as presymptomatic microgliosis precedes overt neuronal atrophy.

Retinal and Hypothalamic Gliosis

In the , a specialized extension of the , gliosis primarily involves Müller cells, glial cells that the retinal layers and neuronal . In , a common complication of , Müller cells undergo reactive gliosis characterized by , , and upregulation of intermediate filaments such as (). This response is triggered by hyperglycemia-induced metabolic and vascular endothelial growth factor () overexpression, leading to impaired glutamate , increased , and contributions to retinal edema and neurodegeneration. A unique aspect of retinal gliosis is its intimate association with vascular pathology; in , gliotic Müller cells interact with dysfunctional retinal blood vessels, exacerbating leakage and ischemia through the release of pro-angiogenic factors. Following retinal detachment, an acute injury where the neurosensory retina separates from the underlying retinal pigment epithelium, GFAP expression markedly increases in the inner retina, particularly in Müller cell processes extending to the inner limiting membrane. This gliotic reaction forms a protective barrier but can also contribute to scar formation, potentially hindering photoreceptor reattachment and visual recovery. Optical coherence tomography (OCT) imaging reveals these gliotic scars as hyper-reflective areas in the inner retinal layers, providing a non-invasive means to assess the extent of reactive changes and their correlation with functional deficits. In the hypothalamus, a critical brain region for metabolic homeostasis, astrogliosis is prominently linked to obesity through chronic low-grade inflammation along the inflammation-obesity axis. A 2025 review highlights that high-fat diet consumption induces hypothalamic astrocyte activation, marked by GFAP upregulation and morphological hypertrophy, which disrupts energy balance regulation and promotes weight gain. This gliosis interferes with leptin signaling, a key hormone for appetite control; reactive astrocytes exhibit impaired leptin receptor function and increased pro-inflammatory cytokine production, leading to central leptin resistance that sustains hyperphagia and insulin insensitivity. Unlike generalized astrocytic responses, hypothalamic gliosis uniquely ties to metabolic dysregulation, as activated astrocytes alter neuronal circuits in the arcuate nucleus, amplifying obesity-related comorbidities. Evidence from magnetic resonance imaging (MRI) studies shows T2 hyperintensities and microstructural changes in the hypothalamus of obese individuals, correlating with gliotic alterations and supporting its role in disease progression.

Therapeutic Targets and Interventions

Enhancing Beneficial Aspects

Strategies to enhance the beneficial aspects of gliosis focus on amplifying its neuroprotective and regenerative roles while optimizing scar formation to facilitate recovery after central nervous system (CNS) injury. One approach involves promoting repair through neurotrophic factors, such as brain-derived neurotrophic factor (BDNF), which supports axonal regeneration, neurogenesis, remyelination, and synaptic plasticity in spinal cord injury (SCI) models. BDNF mimetics delivered via nanofibrous scaffolds have demonstrated improved implant integration and increased neuronal plasticity by enhancing neurite sprouting at tissue-implant interfaces. Similarly, exercise-induced factors boost neurotrophism by normalizing astrocyte morphology, elevating glial fibrillary acidic protein (GFAP) levels, and promoting the release of neurotrophic agents in models of neurodegeneration, thereby mitigating reactive gliosis and supporting dopaminergic neuron protection. Scar optimization represents another key intervention, aiming to partially modulate the glial scar to permit axon passage without complete ablation, preserving its protective barrier function. Scar-homing liposomes, modified with the CAQK peptide to target chondroitin sulfate proteoglycans (CSPGs), deliver docetaxel and BDNF to reduce fibrotic components like neurocan and NG2 while maintaining glial sealing. In rat SCI models, this approach decreased lesion area by approximately 60%, enhanced microtubule stabilization and growth cone activity, and allowed more axons to traverse the scar, as evidenced by biotinylated dextran amine tracing. Functional outcomes included Basso-Beattie-Bresnahan (BBB) scores improving to 12 at 56 days post-injury compared to 8 in controls, alongside restored hindlimb movement. Gene therapy targeting signal transducer and activator of transcription 3 (STAT3) provides compelling evidence for enhancing gliosis-mediated recovery. Sustained STAT3 activation via recombinant adeno-associated virus (rAAV-STAT3) promotes corticospinal tract remodeling by inducing sprouting from lesioned fibers and de novo collaterals from unlesioned ones in mouse pyramidotomy models. This led to significant functional improvements, including enhanced forelimb grasping starting at 5 weeks post-injury (P<0.05) and full restoration of electromyographic responses in 100% of ipsilateral sites by 12 weeks. Clinical trials of stem cell implants further illustrate the potential to modulate early gliosis for better outcomes. Mesenchymal stem cells (MSCs), such as bone marrow-derived or umbilical cord-derived variants, exert paracrine effects that reduce inflammation and astrogliosis while promoting a regenerative microenvironment. In subacute and chronic SCI trials, intrathecal or on-spine implantation of 1-3 × 10^8 autologous BMSCs in 11-50 patients (mostly ASIA A/B) resulted in 27-46% achieving ASIA C or better, with sensory and motor gains attributed to immunomodulation and trophic support. Similarly, a phase I trial of adipose-derived MSCs reported safety and preliminary efficacy in reducing hypersensitivity and improving paralysis scores.

Suppressing Detrimental Aspects

Approaches to suppressing the detrimental aspects of gliosis focus on targeting maladaptive features, such as excessive scar formation and , to promote neural repair while maintaining protective functions of glial cells. For instance, chondroitinase ABC (ChABC), an that degrades chondroitin sulfate proteoglycans (CSPGs)— components of the shown to enhance axonal and functional in models of by creating a more permissive for regeneration. In these studies, intrathecal or intracortical of ChABC reduced CSPG levels and improved locomotor outcomes without completely abolishing the glial response. Anti-inflammatory strategies also play a in curbing that sustains detrimental gliosis. Nonsteroidal drugs (NSAIDs), such as ibuprofen, inhibit RhoA signaling in reactive glia, thereby promoting axonal regeneration and reducing scar-associated inhibition in rat models of spinal cord injury. Similarly, interleukin-1 (IL-1) receptor antagonists, like IL-1ra, suppress reactive astrogliosis by blocking IL-1-mediated of microglia and astrocytes, leading to decreased glial and in models of CNS and . These agents help mitigate the pro-inflammatory that exacerbates tissue during prolonged gliotic responses. The timing of interventions is crucial, as early suppression of gliosis can impair initial protective mechanisms and worsen outcomes, whereas delayed administration—typically weeks post-injury—supports neural plasticity and repair. Genetic or pharmacological inhibition of astrogliosis immediately after injury disrupts barrier formation and increases inflammation, highlighting the need for phased therapeutic strategies. In contrast, late-stage modulation enhances synaptic remodeling and functional recovery in preclinical models. In human applications, minocycline, a tetracycline antibiotic with anti-inflammatory properties, has demonstrated potential in multiple sclerosis (MS) trials by reducing markers of gliosis and neuroinflammation. In the MinoCIS phase III trial, minocycline treatment delayed conversion from clinically isolated syndrome to MS and significantly lowered gadolinium-enhancing lesions on MRI—indicators of active inflammation linked to gliotic processes—compared to placebo over six months. This suggests minocycline's ability to attenuate detrimental glial activation in chronic neuroinflammatory conditions, though larger studies are needed to confirm long-term effects on gliosis specifically.

Emerging Molecular Targets

Recent advances in gliosis research have identified several promising molecular targets for therapeutic intervention, focusing on modulating glial responses in neurodegenerative contexts. TREM2 agonists, which activate the triggering receptor expressed on myeloid cells 2 (TREM2), have shown potential in enhancing microglial phagocytosis of amyloid-beta plaques in Alzheimer's disease (AD) models. Clinical trials initiated in 2024, such as the INVOKE-2 phase 2 study evaluating AL002, a TREM2-activating monoclonal antibody, showed safety in phase 1 but failed to meet the primary endpoint of slowing clinical progression in early AD patients as of November 2024 results. The CX3CL1/CX3CR1 signaling , involving the CX3CL1 (fractalkine) and its receptor CX3CR1 on , represents another emerging for balancing gliotic in neurodegeneration. Modulators that enhance this , such as CX3CR1 agonists, have been shown to promote microglial surveillance and reduce excessive in preclinical models of Parkinson's and AD, preventing synaptic . A 2025 comprehensive highlighted how disrupting this exacerbates gliosis, while targeted modulators could restore , with ongoing preclinical studies exploring small-molecule agonists for clinical . Epigenetic modifications offer novel avenues for reprogramming reactive astrocytes during gliosis, particularly through histone deacetylase (HDAC) inhibitors. These agents have been demonstrated to attenuate astrocytic hypertrophy and GFAP upregulation in spinal cord injury models by altering chromatin accessibility and promoting a pro-regenerative phenotype. Recent in vitro and rodent studies from 2023-2025 indicate that HDAC inhibition reduces scar formation and enhances axonal regrowth, positioning it as a candidate for clinical trials in chronic gliotic conditions.01567-8) Nanotechnology-enabled approaches are advancing targeted delivery to gliotic scar sites, including CRISPR-based systems for GFAP knockdown to mitigate inhibitory astrogliosis. Nanoparticle carriers, such as lipid-based vectors conjugated with gliosis-specific ligands, have achieved precise CRISPR/Cas9 delivery in brain injury models, showing promise for translating to human traumatic CNS injuries without off-target effects.

References

  1. [1]
    Reactive gliosis in traumatic brain injury: a comprehensive review
    In response to tissue trauma, glia cells undergo both morphological and functional changes characterized by the term reactive gliosis, which usually refers to ...
  2. [2]
    Neurotoxic reactive astrocytes are induced by activated microglia
    Jan 18, 2017 · Reactive astrocytes are strongly induced by central nervous system (CNS) injury and disease, but their role is poorly understood.
  3. [3]
    Reactive gliosis in the pathogenesis of CNS diseases - ScienceDirect
    Reactive (astro)gliosis is highly heterogeneous and also context-dependent process that aims at the restoration of homeostasis and limits tissue damage.
  4. [4]
    Interpretation of Gliosis in the Brain and Spinal Cord ... - PubMed
    Apr 14, 2023 · Gliosis, defined as a nonneoplastic reaction (hypertrophy and/or proliferation) of astrocytes and/or microglial cells, is a frequent finding in the central ...Missing: neuroscience review
  5. [5]
    Reactive gliosis in traumatic brain injury: a comprehensive review
    This review summarizes TBI as a complex process, where the resultant effect is severity-, region- and time-dependent and determined by the model of the CNS ...
  6. [6]
    Gliosis | Radiology Reference Article - Radiopaedia.org
    Jan 31, 2024 · Gliosis is a reactive process occurring after some time following most types of central nervous system injuries and is the result of focal proliferation of ...Missing: neuroscience | Show results with:neuroscience
  7. [7]
    Glia - an overview | ScienceDirect Topics
    Rudolf Virchow first described glia in 1846, considering them as connective ... astrocytes and glial scar formation under pathologic conditions including CNS ...
  8. [8]
    An Early History of Neuroglial Research: Personalities - MDPI
    The passive part of the nervous tissue has been further conceptualised by Rudolf Virchow, who introduced the notion of neuroglia as a connective tissue of the ...
  9. [9]
    Translation and comments on Pío del Río-Hortega 1919 ... - PubMed
    The "Big-Bang" for modern glial biology: Translation and comments on Pío del Río-Hortega 1919 series of papers on microglia · Authors · Affiliations.Missing: classification | Show results with:classification
  10. [10]
    Reactive astrocyte nomenclature, definitions, and future directions
    Feb 15, 2021 · From the early 20th century up to the 1980s, the morphological appearance of astrocytes was the only readout of their role in neuropathology.
  11. [11]
    Cien Años de Microglía: Milestones in a Century of Microglial ...
    Microglia were first described by the Spanish researcher Pío del Río-Hortega in 1919. · Río-Hortega's discoveries identified and defined the three types of glial ...
  12. [12]
    Toxicologic Pathology Forum Opinion: Interpretation of Gliosis in the ...
    Apr 14, 2023 · Gliosis, defined as a nonneoplastic reaction (hypertrophy and/or proliferation) of astrocytes and/or microglial cells, is a frequent finding in the central ...
  13. [13]
    Astrocytes: CNS Functions in Health & Disease Review
    May 26, 2023 · Astrocytes contribute to the formation and maintenance of the blood-brain barrier. They provide structural support and influence the transport ...
  14. [14]
    Astrocytes in human central nervous system diseases: a frontier for ...
    Oct 13, 2023 · Astroglia are a broad class of neural parenchymal cells primarily dedicated to homoeostasis and defence of the central nervous system (CNS).
  15. [15]
    Astrocyte metabolism and signaling pathways in the CNS - Frontiers
    Protoplasmic astrocytes have various functions, including modulation of synaptic function, clearance of glutamate, regulation of blood flowrate, and ...
  16. [16]
    Microglia Function in the Central Nervous System During Health and ...
    Microglia are resident cells of the brain that regulate brain development, maintenance of neuronal networks, and injury repair.
  17. [17]
    Microglia: Structure, function and origin | Kenhub
    Microglia represent the only form of native active immunity in the CNS, serving as the primary defense and preservation mechanism.
  18. [18]
    The ins and outs of microglial cells in brain health and disease
    Microglia are the brain's resident macrophages that play pivotal roles in immune surveillance and maintaining homeostasis of the Central Nervous System (CNS).
  19. [19]
    Functions and dysfunctions of oligodendrocytes in ... - Frontiers
    Oligodendrocytes (OLs), the myelinating cells of the central nervous system (CNS), enable rapid transmission of electrical signals and provide metabolic and ...
  20. [20]
    Reactive gliosis and the multicellular response to CNS damage and ...
    'Reactive gliosis' will refer not only to microglia and astroglia, but also to glial cells that have come to be known as NG2-positive oligodendrocyte progenitor ...
  21. [21]
    Roles of Ependymal Cells in the Physiology and Pathology of the ...
    Ependymal cells play key roles in mammalian CNS development and normal physiological processes by controlling the production and flow of cerebrospinal fluid ( ...
  22. [22]
    Ependymal cells undergo astrocyte‐like reactivity in response to ...
    May 4, 2024 · Ependymal cells are unique glia that regulate local cerebrospinal fluid (CSF) microcirculation and form a dynamic interface between the ...3.1 Ependymal Cells Assume A... · 3.2 Ependymal Gfap, Stat1... · 3.3 Ependymal Gfap And Stat1...
  23. [23]
    Histology, Glial Cells - StatPearls - NCBI Bookshelf - NIH
    Glial cells are the most abundant cells in the central nervous system. The most notable glial cells include oligodendrocytes, Schwann cells, astrocytes, ...
  24. [24]
    Astrocyte Reactivity and Reactive Astrogliosis: Costs and Benefits
    Oct 1, 2014 · [From Pekny and Pekna (176).] B: reactive astrocytes in an epileptic focus from a patient who suffered from pharmacologically intractable ...
  25. [25]
    Molecular dissection of reactive astrogliosis and glial scar formation
    This article reviews developments in the signaling mechanisms that regulate specific aspects of reactive astrogliosis and highlights the potential to identify ...
  26. [26]
    Signaling Pathways in Reactive Astrocytes, a Genetic Perspective
    Jan 14, 2011 · In this review, we limit our discussion primarily to what molecular mechanisms regulate astrocyte activation. Many cell types including neurons ...
  27. [27]
    Detrimental Effects of HMGB-1 Require Microglial-Astroglial Interaction
    Aug 27, 2019 · High Mobility Group Box 1 (HMGB1) is an ubiquitous nuclear protein that is implicated in the maintenance of chromatin structure (Ellwood et al., ...
  28. [28]
    Pten Loss Causes Hypertrophy and Increased Proliferation of ...
    Nov 1, 2004 · This study shows cell-type dependent effects of Pten loss in the adult brain, including increased astrocyte proliferation that may render astroglial cells ...
  29. [29]
    MiR-29a Knockout Aggravates Neurological Damage by ... - Frontiers
    MiR-29a-5p Knockout Promoted Astrocyte Proliferation and Increased the Release of Glutamate in Basal Ganglia of Rats After MCAO. After knockout of miR-29a-5p, ...
  30. [30]
    Minocycline reduces reactive gliosis in the rat model of hydrocephalus
    Dec 5, 2012 · The mechanism might operate through inhibition of astrogliosis and glial scar formation thereby decreasing the damage to periventricular white ...<|control11|><|separator|>
  31. [31]
    Endothelin-1 Regulates Astrocyte Proliferation and Reactive Gliosis ...
    We show that ET-1 levels are upregulated in demyelinated lesions within 5 d after LPC injection, together with enhanced astrocyte proliferation, GFAP ...
  32. [32]
    Astrocyte Reactivity and Reactive Astrogliosis: Costs and Benefits
    Oct 1, 2014 · Reactive astrogliosis, a response of astrocytes seen in a multitude of neurological disorders, can be defined as constitutive, graded, ...<|control11|><|separator|>
  33. [33]
    High susceptibility to cerebral ischemia in GFAP-null mice - PubMed
    Laser-Doppler flowmetry revealed that the GFAP-null mice had a more extensive and profound decrease in cortical cerebral blood flow within 2 minutes after MCAO ...Missing: percentage | Show results with:percentage
  34. [34]
    Pathophysiological Responses and Roles of Astrocytes in Traumatic ...
    Emerging evidence suggests that astrocytes elicit beneficial and detrimental roles in the pathogenesis of TBI, including neuroinflammation, brain edema, ...5.1. Neurogenesis And... · 5.2. Bbb Disruption And... · 5.3. Cytotoxic Edema
  35. [35]
  36. [36]
    Tumor Necrosis Factor Alpha: A Link between Neuroinflammation ...
    Neuroinflammation and excitotoxicity have key roles as triggers and sustainers of the neurodegenerative process and thus, elevated levels of TNF-α have been ...
  37. [37]
    Conditional ablation of Stat3 or Socs3 discloses a dual ... - PubMed
    Jun 18, 2006 · These results suggest that Stat3 is a key regulator of reactive astrocytes in the healing process after SCI, providing a potential target for intervention.
  38. [38]
    Epigenetic Regulation of Astrocyte Function in Neuroinflammation ...
    Therefore, increased histone acetylation might affect reactive astrogliosis through reducing STAT3 signaling, similar to that seen with DNA methylation during ...
  39. [39]
    STAT3 is a Critical Regulator of Astrogliosis and Scar Formation ...
    Jul 9, 2008 · STAT3 signaling in astrocytes is important in regulating scar formation and that disrupted scar formation exacerbates inflammation after SCI.
  40. [40]
    Microglia in neurodegenerative diseases: mechanism and potential ...
    Sep 22, 2023 · In 1919, Spanish researcher Pı´o del Rı´o-Hortega discovered a new type of glial cell based on his invention of a novel method to stain the ...<|separator|>
  41. [41]
    Microglial M1/M2 polarization and metabolic states - PMC
    This review examines the polarization states of microglia and their relationship to mitochondrial metabolism.
  42. [42]
    Analyzing microglial phenotypes across neuropathologies
    Oct 8, 2021 · Upon activation, microglia rapidly change their morphology within a few minutes. Typically, the microglial somata appear bigger and the arms ...Missing: gliosis timeline
  43. [43]
    What are activated and reactive glia and what is their role in ...
    These states were correspondingly named “M1” and “M2”. In this model, M1-activated macrophages produce more iNOS and typically pro-inflammatory NFkB ...Missing: paper | Show results with:paper
  44. [44]
    Regulation of Microglial Functions by Purinergic Mechanisms in the ...
    It has been known for a long time that neuronal damage leads to the activation of ramified microglia through several steps transforming them by multiple factors ...
  45. [45]
    Microglia activation in central nervous system disorders: A review of ...
    Mar 7, 2023 · Microglia can remain chronically activated for years after the acute inflammatory process has subsided. In patients with TBI, microglia ...
  46. [46]
    Different phenotypes of microglia in animal models of Alzheimer ...
    Oct 8, 2022 · Microglial activation has been considered as heterogeneous, with categorization into the M0, M1, and M2 phenotypes involved in surveillance, pro ...
  47. [47]
    The Impact of the CX3CL1/CX3CR1 Axis in Neurological Disorders
    Oct 13, 2020 · Recently, a chemokine, fractalkine (CX3CL1), and its unique receptor (CX3CR1) were shown to be able to regulate the activation of microglia to ...Missing: gliosis | Show results with:gliosis
  48. [48]
    Modulation of the Purinergic P2X7 Receptor Attenuates ...
    May 2, 2007 · Thus, activation of P2X7R in microglia in inflammatory conditions is associated with enhancement of neuronal damage, an effect that is ...
  49. [49]
    Mechanisms Underlying Interferon-γ-Induced Priming of Microglial ...
    Microglial priming and enhanced reactivity to secondary insults cause substantial neuronal damage and are hallmarks of brain aging, traumatic brain injury ...
  50. [50]
    TREM2 dependent and independent functions of microglia in ...
    Dec 23, 2022 · Acute Trem2 reduction triggers increased microglial phagocytosis, slowing amyloid deposition in mice. Proc Natl Acad Sci U S A. 2021;118(27): ...Microglia: From Cns... · Trem2 Sustains Microglial... · Trem2 Controls Microglia...
  51. [51]
    CD200-CD200R Pathway: A Regulator of Microglial Polarization in ...
    Nov 7, 2024 · The CD200-CD200R signaling pathway inhibits M1 polarization by suppressing pro-inflammatory signals such as NF-κB and promotes M2 polarization ...
  52. [52]
    CX3CL1/CX3CR1 Signaling Mediated Neuroglia Activation Is ...
    Jan 17, 2024 · CX3CL1/CX3CR1 signaling has been documented to mediate the microglia activation and gliosis reaction during neurodegeneration. We intend to ...
  53. [53]
    Co-stimulation with IL-1β and TNF-α induces an inflammatory ...
    Nov 15, 2019 · Co-stimulation with IL-1β and TNF-α induces an inflammatory reactive astrocyte phenotype with neurosupportive characteristics in a human ...
  54. [54]
    Reciprocal modulation between microglia and astrocyte in reactive ...
    As two major cellular populations of reactive gliosis, microglia and astrocytes can activate each other and have a tightly reciprocal modulation during the GS ...
  55. [55]
    Astrocytic phagocytosis is a compensatory mechanism for microglial ...
    Sep 22, 2020 · Astrocytes possess phagocytic machinery and have the potential to compensate for microglia with dysfunctional phagocytic activity.<|separator|>
  56. [56]
    Phagocytic Roles of Glial Cells in Healthy and Diseased Brains - PMC
    Microglia exhibit increased immune responses, including phagocytosis, following various stimuli and aging, but astrocytes appear to lose their phagocytic ...
  57. [57]
    Mouse Astrocytes Promote Microglial Ramification by Releasing ...
    Jul 9, 2020 · Blocking astrocyte-derived TGF-β decreased microglial ramification. Astrocytes induced the formation of protuberances on branches of microglia ...
  58. [58]
    TGFβ produced by IL-10 re-directed Astrocytes Attenuates Microglial ...
    Here we show that IL-10 re-directed astrocytes regulate the activation of microglia in a Transforming growth factor (TGF)-β dependent manner.Missing: shift | Show results with:shift
  59. [59]
    Microglia activation triggers astrocyte-mediated modulation ... - PNAS
    Dec 13, 2011 · Our data indicate that, on activation, microglia cells rapidly release small amounts of ATP, and astrocytes, in turn, amplified this release.
  60. [60]
    How astrocytic ATP shapes neuronal activity and brain circuits
    ATP release from astrocytes can activate P2Y12Rs, the receptors in charge of microglial targeted motility to synapses and neuronal somata [53, 54, 55•]. As ...
  61. [61]
    Microglia are an essential component of the neuroprotective scar ...
    Jan 31, 2019 · A microglial scar forms at the astrocyte–immune cell interface. It was recently proposed that cytokines released by activated microglia in ...
  62. [62]
    Continual Deletion of Spinal Microglia Reforms Astrocyte Scar ...
    Jun 27, 2022 · Recent advances have found that microglia are necessary for the activation of astrocytes in response to SCI to form the compact astrocyte scar ...
  63. [63]
    A1 reactive astrocytes and a loss of TREM2 are associated with an ...
    Jul 25, 2020 · A1 astrocyte conditioned media decreases TREM2 protein in mouse primary microglia culture. ... TREM2 deficiency exacerbates tau pathology ...
  64. [64]
    Triggering Receptor Expressed on Myeloid Cells 2 (TREM2 ...
    Feb 25, 2015 · Microglia deficient in TREM2 showed less activation and failed to phagocytose OGD-exposed neurons. Primary mixed cultures of neurons, astrocytes ...
  65. [65]
    Microglia amplify inflammatory activation of astrocytes in ...
    May 5, 2017 · Cell culture models demonstrate that Mn potentiates inflammatory gene expression in microglia following lipopolysaccharide (LPS)/cytokine ...
  66. [66]
    Crosstalk Between Astrocytes and Microglia: An Overview - Frontiers
    Jul 15, 2020 · Microglia and astrocytes respond to neuronal injury with programs that include proliferation, morphological alterations, mediator production, ...
  67. [67]
  68. [68]
    NG2 Is a Major Chondroitin Sulfate Proteoglycan Produced after ...
    Apr 1, 2002 · NG2 expression results from a multicellular response to injury, including both reactive macrophages and oligodendrocyte progenitors; astrocytes ...
  69. [69]
    Chronic oligodendrocyte injury in central nervous system pathologies
    Nov 19, 2022 · Injury to myelin-forming oligodendrocytes (OL) in chronic neurological diseases (eg multiple sclerosis) ranges from sublethal to lethal.
  70. [70]
    An inhibitor of chondroitin sulfate proteoglycan synthesis promotes ...
    Apr 26, 2016 · We introduce a novel CSPG synthesis inhibitor to reduce CSPG content and find rescued process outgrowth from OPCs in vitro and accelerated remyelination.
  71. [71]
    Differentiation block of oligodendroglial progenitor cells as a cause ...
    In summary, we provide evidence that a differentiation block of oligodendroglial progenitors is a major determinant of remyelination failure in chronic multiple ...<|separator|>
  72. [72]
    Traumatic Brain Injury-Induced Ependymal Ciliary Loss Decreases ...
    Concussive brain injury leads to rapid and protracted loss of ventricular ependymal cilia without overt ependymal cell loss. The ventricular system of the brain ...Missing: proliferation | Show results with:proliferation
  73. [73]
    Therapeutic strategies to recover ependymal barrier after ... - Frontiers
    The recovery and regeneration of ependyma after damage are key to stabilizing neuroinflammatory and neurodegenerative processes that are critical during early ...Missing: breaches | Show results with:breaches
  74. [74]
    Glial Remodeling in the Ventricular–Subventricular Zone and ... - MDPI
    After two weeks of age, hydrocephalus leads to hemosiderin accumulation, nodular gliosis, ependymal cell loss, and the formation of subependymal rosettes in the ...
  75. [75]
    Reactive Bergmann glia play a central role in spinocerebellar ataxia ...
    Using human SCA autopsy samples we have discovered that Bergmann glia—the radial glia of the cerebellum, which form intimate functional connections with ...
  76. [76]
    Roles of Ependymal Cells in the Physiology and ... - PubMed Central
    Ependymal cells play key roles in mammalian CNS development and normal physiological processes by controlling the production and flow of cerebrospinal fluid ( ...
  77. [77]
    Ependymal cells and neurodegenerative disease - Oxford Academic
    Nov 4, 2022 · These findings suggest that DNA damage in the ependymal cell lining may be an early molecular event post-mTBI and may precede neuropathological ...Missing: seal breaches
  78. [78]
    Mechanically Induced Reactive Gliosis Causes ATP-Mediated ...
    In this study, the mechanical properties of cultured cortical astrocytes are measured following a mechanical stretch injury that induces reactive gliosis.Missing: acute | Show results with:acute
  79. [79]
    Activation of Extracellular Signal-Regulated Kinase by Stretch ...
    Mar 15, 2003 · These findings demonstrate for the first time that ATP released by mechanical injury is one of the signals that triggers ERK activation and ...
  80. [80]
    Lactate Promotes Reactive Astrogliosis and Confers Axon Guidance ...
    Aug 21, 2020 · During cerebral ischemia, brain lactate concentration increases, and astrogliosis is triggered. Herein, we investigated lactate's role in ...Missing: hypoxia ROS
  81. [81]
    Astrogliosis and glial scar in ischemic stroke - ScienceDirect.com
    At the same time, reactive astrocytes release neuroprotective factors, ultimately relieving brain injury. In the late stage of ischemia, reactive astrocytes ...
  82. [82]
    Crosstalk Between the Oxidative Stress and Glia Cells After Stroke
    Astrocytes and microglia are rapidly activated after stroke, generating large amounts of ROS via mitochondrial and NADPH oxidase pathways, causing oxidative ...
  83. [83]
    ASTROGLIOSIS INVOLVES ACTIVATION OF RIG-LIKE SIGNALING ...
    Our findings identify a role for RLR signaling and type I IFN in regulating astrocyte innate immune responses after SCI.
  84. [84]
    Viral CNS infections: role of glial pattern recognition receptors in ...
    Jun 20, 2012 · RNA and DNA viruses trigger broad activation of glial cells including microglia and astrocytes, eliciting the release of an array of mediators ...
  85. [85]
    The S100B Protein: A Multifaceted Pathogenic Factor More Than a ...
    These observations confirmed a putative role played by S100B in acute brain injury pathogenic processes. Likewise, treatment with a neutralizing anti-S100B ...
  86. [86]
    Role of damage-associated molecular patterns in the pathogenesis ...
    Jul 13, 2025 · TBI is a common cause of DAI, and DAI is the most common form of diffuse brain injury [49]. DAI results from shear forces on axons caused by ...
  87. [87]
    DAMPs and RAGE Pathophysiology at the Acute Phase of Brain Injury
    Later, at the subacute phase, TLRs and RAGE activation will polarize microglial cells and infiltrating leukocytes toward a pro-inflammatory phenotype, termed M1 ...
  88. [88]
    RAGE is a key cellular target for Aβ-induced perturbation in ...
    RAGE is an important cellular cofactor for amyloid β-peptide (Aβ)-mediated cellular perturbation relevant to the pathogenesis of Alzheimer's disease (AD).
  89. [89]
    Targeting neuroinflammation in Alzheimer's disease - PMC
    Nov 3, 2016 · Reactive gliosis is now considered a key abnormality in the AD brain. It has been demonstrated that reactive astrocytes surround both Aβ plaques ...
  90. [90]
    The S100B/RAGE Axis in Alzheimer's Disease - PubMed Central - NIH
    A soluble form of RAGE can also be generated by proteolysis. S100B, AGEs Aβ oligomers, and TTR bind to RAGE V domain. Aβ aggregates binds to RAGE C1 domain.
  91. [91]
    Tau hyperphosphorylation and increased BACE1 and RAGE levels ...
    Our findings indicate that PPARβ/δ deficiency results in cognitive impairment associated with enhanced inflammation, astrogliosis and tau hyperphosphorylation ...<|control11|><|separator|>
  92. [92]
    MHCII Is Required for α-Synuclein-Induced Activation of Microglia ...
    Jun 5, 2013 · Accumulation of α-synuclein (α-syn) in the brain is a core feature of Parkinson disease (PD) and leads to microglial activation, production of ...
  93. [93]
    Glial dysfunction in the pathogenesis of α-synucleinopathies
    Accumulating experimental evidence in PD models suggests that astrogliosis and microgliosis act as important mediators of neurodegeneration playing a pivotal ...
  94. [94]
    α-Synuclein-activated microglia are implicated in PD pathogenesis
    Feb 14, 2022 · “Our findings point strongly toward α-synuclein oligomers being the main toxic participants in PD,” concludes Buttini. “We uncovered evidence ...
  95. [95]
    Hypothalamic inflammation and gliosis in obesity - PMC
    High-fat diet feeding is associated with hypothalamic inflammation, reactive gliosis, and neuron injury in rodents. Interventions that increase or decrease ...Missing: 2024 | Show results with:2024
  96. [96]
    Hypothalamic inflammation in obesity and metabolic disease - JCI
    Jan 3, 2017 · There is growing evidence that consumption of a high-fat, high carbohydrate diet is associated with hypothalamic deregulation in the human brain ...
  97. [97]
    The Significance of Hypothalamic Inflammation and Gliosis for the ...
    Emerging translational evidence supports that hypothalamic inflammation and gliosis contribute to obesity and its comorbidities in humans, as seen in rodent ...
  98. [98]
    Hypothalamic neuronal-glial crosstalk in metabolic disease - Nature
    Oct 4, 2024 · Hypothalamic gliosis associated with high-fat diet feeding is reversible in mice: a combined immunohistochemical and magnetic resonance ...
  99. [99]
    The role of the complement system in Multiple Sclerosis: A review
    In this review, we evaluated the possible role of complement activation in multiple sclerosis (MS) with a focus in progressive MS.
  100. [100]
    Multiple sclerosis and myelin basic protein
    Dec 10, 2021 · MBP has long been studied as a factor in the pathogenesis of the autoimmune neurodegenerative disease multiple sclerosis (MS).
  101. [101]
    Complement is activated in progressive multiple sclerosis cortical ...
    Jun 22, 2016 · Complement is activated in the MS cortical grey matter lesions in areas of elevated numbers of complement receptor-positive microglia.
  102. [102]
    Complement Activation Is Associated With Disease Severity in ...
    Feb 14, 2024 · Complement C3 on microglial clusters in multiple sclerosis occur in chronic but not acute disease: implication for disease pathogenesis. Glia.
  103. [103]
    The Contribution of Complement System in Multiple Sclerosis ...
    Apr 16, 2025 · Our review examines the role of the complement system in multiple sclerosis (MS), an autoimmune disease causing CNS myelin destruction.The Complement System · Complement System in... · Classical Pathway of...
  104. [104]
    Aberrant DNA methylation profile exacerbates inflammation and ...
    Jan 14, 2020 · An exhaustive study of DNA methylation might contribute towards new pharmacological interventions in MS by use of epigenetic drugs.<|control11|><|separator|>
  105. [105]
    DNA methylation changes in glial cells of the normal-appearing ...
    We aimed to examine DNA methylation, a stable epigenetic mark of genome activity, in glial cells to capture relevant molecular changes underlying MS ...Missing: modifications | Show results with:modifications
  106. [106]
    The role of transcriptional and epigenetic modifications in ...
    Sep 20, 2024 · Histone modifications, including acetylation, methylation, and ubiquitination, affect chromatin accessibility and, as a consequence, regulate ...
  107. [107]
    Microglia/macrophage polarization dynamics in white matter after ...
    Aug 14, 2013 · ... days after TBI and peaked by 3 to 5 days post injury. The majority of M2-type genes began to decrease at 7 days and returned to pre-injury ...
  108. [108]
    Reactive gliosis in traumatic brain injury: a comprehensive review
    Feb 28, 2024 · This review summarizes TBI as a complex process, where the resultant effect is severity-, region- and time-dependent and determined by the model of the CNS ...
  109. [109]
    Moving beyond the glial scar for spinal cord repair - Nature
    Aug 28, 2019 · Pharmacological suppression of CNS scarring by deferoxamine reduces lesion volume and increases regeneration in an in vitro model for ...
  110. [110]
    Tackling the glial scar in spinal cord regeneration - PubMed Central
    ... detrimental roles of glial and stromal cells. ... Harmful and beneficial effects of inflammation after spinal cord injury: potential therapeutic implications.
  111. [111]
    Reactive Gliosis and the Multicellular Response to CNS Damage ...
    Jan 22, 2014 · This Review examines and reviews the multiple cell types involved in, and contributing to, different types of CNS insults.
  112. [112]
    Glial fibrillary acidic protein elevations relate to neuroimaging ... - NIH
    Our study confirms GFAP as a promising marker of brain injury in patients with acute mTBI. A combination of various biomarkers linked to different ...
  113. [113]
    Association of CSF and Serum Neurofilament Light and Glial ...
    This study provides Class I evidence that higher serum NF-L and GFAP are associated with worse neurological outcome after acute SCI.
  114. [114]
    Microglial Phenotypes and Functions in Multiple Sclerosis - PMC
    Active demyelination is associated with inflammation and reactive microgliosis (Lassmann 2014). New focal lesions can appear at any site of the brain and spinal ...
  115. [115]
    Profiling of microglia nodules in multiple sclerosis reveals propensity ...
    Feb 23, 2024 · We show that microglia nodules are associated with more severe MS pathology. Compared to microglia nodules in stroke, those in MS show enhanced expression of ...
  116. [116]
    The Role of Astrocytes in Multiple Sclerosis - PMC - PubMed Central
    Conclusion. Astrocytes play an instrumental role in the formation of MS lesions through a multitude of functional changes associated with their activation.
  117. [117]
    A Therapeutic Link between Astrogliosis and Remyelination in a ...
    Jan 3, 2018 · The inability of oligodendrocytes in MS lesions to successfully remyelinate damaged axons suggests that some aspect of the lesion environment, ...Missing: gliosis | Show results with:gliosis
  118. [118]
    Myeloid lineage C3 induces reactive gliosis and neuronal stress ...
    Apr 12, 2025 · We find that C3 expression in the early inflammatory stage of EAE defines disease-associated glial subtypes characterized by increased expression of genes.
  119. [119]
    Multiple Sclerosis Pathology - PMC - PubMed Central - NIH
    Demyelination and neurodegeneration in the MS brain is associated with a profound astroglia reaction, forming a dense glial scar in long-standing established ...
  120. [120]
    The impact of regional astrocyte interferon-γ signaling during ...
    Jun 12, 2020 · Here, we demonstrate that IFNγ signaling in regional astrocytes induces the iP and promotes protection of the CNS during chronic autoimmunity.
  121. [121]
    Astrogliosis in multiple sclerosis and neuro-inflammation - Frontiers
    During neuro-inflammation, astrocytes become reactive. They undergo morphological and molecular changes named “astrogliosis” driving the conversion from acute ...
  122. [122]
    Central nervous system macrophages in progressive multiple sclerosis
    Feb 10, 2022 · Multiple sclerosis (MS) is a chronic immune-mediated disease characterized by inflammation, demyelination, gliosis, and neurodegeneration in the ...<|control11|><|separator|>
  123. [123]
    Microglia and Astrocyte Dysfunction in Parkinson's Disease - PMC
    Recent studies have identified that PD-associated genes are expressed in glial cells as well as neurons and have crucial roles in microglia and astrocytes.
  124. [124]
    Microglia in Alzheimer's disease - PMC - PubMed Central - NIH
    Proliferation and activation of microglia in the brain, concentrated around amyloid plaques, is a prominent feature of Alzheimer's disease (AD).
  125. [125]
    Microglia and Astrocytes in Alzheimer's Disease - PubMed Central
    Aβ plaques create a unique environment that facilitates local tau seeding in dystrophic axons, leading to the propagation and formation of neuritic plaque (NP) ...
  126. [126]
    The Role of Microglia and Astrocytes in Huntington's Disease - PMC
    Microglia and astrocytes are critical in regulating neuronal activity and maintaining an optimal milieu for neuronal function.
  127. [127]
    Bridging systemic metabolic dysfunction and Alzheimer's disease
    May 28, 2025 · Metabolic disorders, including insulin resistance (IR), obesity, and diabetes mellitus, have been strongly linked to an increased risk of AD, ...
  128. [128]
    Müller Cells and Diabetic Retinopathy - PMC - PubMed Central
    Abstract. Müller cells are one of the primary glial cell types found in the retina and play a significant role in maintaining retinal function and health.
  129. [129]
    The role of retinal Müller cells in diabetic retinopathy and related ...
    This article reviews the mechanism of RMCs involvement in DR and the progress in related treatments.Abstract · Introduction · RMCs and DR · Role of RMCs as a target of...
  130. [130]
    Contribution of Müller Cells in the Diabetic Retinopathy Development
    Increasing evidence indicates that Müller cells are affected in diabetic retinopathy and may contribute to the severity of the disease.
  131. [131]
    Glial fibrillary acidic protein increases in Müller cells after retinal ...
    In normal retinas and in attached retinal regions of eyes with retinal detachment, GFAP was detected only in the end feet of the Müller cells as 10 nm diameter ...
  132. [132]
    Differential Response of Müller Cells and Microglia in a Mouse ...
    In this study, GFAP expression, the activation marker of Müller glial cells, increased from the inner limiting membrane of the detached region to the ONL until ...
  133. [133]
    Restoration of retinal morphology and residual scarring after ...
    Apr 5, 2013 · After 2 months, there was a significant central gliotic scar, which appeared hyper-reflective on OCT. Glial origin of the retinal scar ...
  134. [134]
    Update on Hypothalamic Inflammation and Gliosis - PubMed - NIH
    Jan 8, 2025 · The most recent translational studies support the link between hypothalamic inflammation and gliosis and obesity pathogenesis in humans.
  135. [135]
    Higher body mass index is linked to altered hypothalamic ... - Nature
    Nov 22, 2019 · Animal studies suggest that obesity-related diets induce structural changes in the hypothalamus, a key brain area involved in energy ...
  136. [136]
    The contribution of astrocytes to obesity-associated metabolic ...
    The hypothalamic inflammation associated with excessive weight is also true for humans: in the MRI study, a T2 hyperintensity in the hypothalamus imaging was ...<|control11|><|separator|>
  137. [137]
    Chondroitinase ABC Promotes Sprouting of Intact and Injured Spinal ...
    Degradation of CSPGs using the enzyme chondroitinase ABC (ChABC) can promote functional recovery after spinal cord injury. However, the mechanisms underlying ...
  138. [138]
    Chondroitinase ABC in spinal cord injury: advances in delivery ...
    Jun 8, 2025 · Chondroitinase ABC (ChABC), a bacterial enzyme capable of degrading CSPGs ... ChABC alone promoted a 20% increase in axonal regeneration in ...
  139. [139]
    Nonsteroidal Anti-Inflammatory Drugs Promote Axon Regeneration ...
    Apr 11, 2007 · Ibuprofen promotes corticospinal and serotonergic axonal growth in rat spinal cord with dorsal hemisection. To the extent that RhoA activation ...
  140. [140]
    Differential Neuroprotective Effects of Interleukin-1 Receptor ...
    Jan 26, 2018 · We hypothesized that the interleukin-1 receptor antagonist (IL1RA) protects neuronal populations and suppresses apoptosis and gliosis after ...
  141. [141]
    Astrogliosis in the Neonatal and Adult Murine Brain Post-Trauma
    May 15, 1997 · (1996) reported recently that the administration of an IL-1 receptor antagonist protein substantially reduced the number of reactive astrocytes ...
  142. [142]
    Inhibition of Stat3‐mediated astrogliosis ameliorates pathology in an ...
    Modulation of Stat3‐mediated reactive astrogliosis in an Alzheimer's disease mouse model attenuates pathology, enhances amyloid clearance and protects from ...Missing: exacerbation | Show results with:exacerbation
  143. [143]
    Reversal of Established Traumatic Brain Injury-Induced, Anxiety ...
    We examined the efficacy of an anti-inflammatory treatment in decreasing anxiety-like behavior and reactive gliosis when introduced at 1 month after injury.
  144. [144]
    Trial of Minocycline in a Clinically Isolated Syndrome of Multiple ...
    Jun 1, 2017 · The risk of conversion from a clinically isolated syndrome to multiple sclerosis was significantly lower with minocycline than with placebo over 6 months but ...Missing: gliosis | Show results with:gliosis
  145. [145]
    Minocycline reduces gadolinium-enhancing magnetic ... - PubMed
    Minocycline reduces gadolinium-enhancing magnetic resonance imaging lesions in multiple sclerosis. Ann Neurol. 2004 May;55(5):756. doi: 10.1002/ana.20111.Missing: markers trials<|control11|><|separator|>