Summary
Progressive multiple sclerosis (PMS) is an immune-initiated neurodegenerative condition that lacks effective therapies. While peripheral immune infiltration is a hallmark of relapsing-remitting MS (RRMS), PMS is associated with chronic, tissue-restricted inflammation and disease associated reactive glial states. The effector functions of disease associated microglia, astrocytes, and oligodendrocyte lineage cells are beginning to be defined, and recent studies have made significant progress in uncovering their pathologic implications. In this review, we discuss the immune-glia interactions that underlie demyelination, failed remyelination, and neurodegeneration with a focus on PMS. We highlight the common and divergent immune mechanisms by which glial cells acquire disease associated phenotypes. Finally, we discuss recent advances that have revealed promising novel therapeutic targets for the treatment of PMS and other neurodegenerative diseases.
Keywords: Progressive multiple sclerosis, astrocytes, microglia, oligodendrocytes, reactive glia, remyelination, neuroprotection, complement, chronic inflammation, inhibitory factors
Graphical Abstract
Progressive multiple sclerosis (PMS) is a neurodegenerative condition with chronic inflammation and glial cell pathology. In this review, Garton et al. discuss immune-glia interactions in PMS, highlighting potential therapeutic targets for PMS and other neurodegenerative diseases.
Introduction
Multiple sclerosis (MS) is a disease characterized by autoimmune neuroinflammation resulting in demyelination and axonal injury and culminating in neurodegeneration and progressive disability. Discrete relapses, driven by peripheral immune cells migrating into the CNS, tend to occur in the early phases of MS, termed ‘relapsing-remitting’ MS (RRMS). RRMS often occurs over the background of a slow progression of symptoms in the absence of disease flares (‘secondarily progressive’; SPMS). In the primary progressive form (PPMS), patients develop a gradually worsening disease without any remissions. Despite recent discussion about whether this triad system of classification should be transitioned into a more diffuse spectrum of disease1, it is clear that the two ends of the spectrum are not treatable with similar strategies. As we will discuss below, there is a paucity of treatments for the two progressive forms of MS (hereafter termed PMS) in part due to the compartmentalized nature of pathology in PMS. To uncover new therapeutic strategies for PMS, it is essential to investigate the resident CNS glial cells and their roles in perpetuating the chronic inflammatory environment that characterizes PMS. It should be noted that adaptive immunity has recently been suggested to also contribute to the inflammatory environment in PMS, with resident effector memory T cells and B cell infiltration being associated with disease severity2–6; a comprehensive discussion of adaptive immunity falls outside the scope of this review, however. Here we will discuss current findings with regards to demyelination, glial activation and neurotoxicity, and the challenges the field has faced when it comes to treating PMS.
Immune-glia interactions contribute to demyelination and failed remyelination in MS
Myelin is a lipoprotein sheath produced by oligodendrocytes (OLs) which wraps axons, speeding conduction and providing metabolic and trophic support to neurons. The immune-mediated destruction of myelin, a hallmark of MS, therefore has direct consequences on neuronal function and integrity. Depending on the anatomic structures involved, demyelinating lesions can be asymptomatic or lead to diverse and disabling focal symptoms of MS. Over time, chronically demyelinated neurons are vulnerable to axonal injury and metabolic stress, contributing to neurodegeneration in PMS7,8. Oligodendrocyte precursor cells (OPCs) are widely distributed glia with the capacity to differentiate into myelinating OLs in addition to having intrinsic homeostatic functions such as interactions with neurons and the vasculature9–13. However, while some OPC-mediated remyelination occurs, especially in the early stages of MS, it is limited in later PMS14–17. Furthermore, there is substantial heterogeneity in remyelination between individuals and even between lesions in a single person with MS (PwMS), suggesting that dynamic features of the local environment enhance or inhibit remyelination. Myelination-promoting therapies could profoundly improve patient quality of life and impact disease progression, and to this end significant effort has been devoted to defining mechanisms behind demyelination and failed remyelination in MS. Immune-glia interactions, initiated by peripheral immune infiltration and perpetuated by reactive glial phenotypes, drive demyelination and failed remyelination18. In this section we will discuss how peripheral immune molecules act on OL lineage cells and contribute to their death and dysfunction. We also review recent literature describing inflammatory OLs (iOLs) and OPCs (iOPCs) in MS and models of inflammatory demyelination.
Demyelination with failed or limited remyelination is a common feature of MS lesions, which otherwise have variable amounts of inflammation, axonal injury, OL loss, and reactive gliosis based on lesion age and activity19–21. Based on these characteristics, lesions can be subdivided into three categories. The first are acute active lesions, which have breakdown of the BBB, perivascular lymphocyte infiltration (mostly cytotoxic T cells), abundant microglia and macrophages throughout the lesion, and variable axonal injury. Chronic active lesions, in contrast, have a hypocellular and gliotic core with a rim of activated myeloid cells. Finally, inactive lesions are sparsely cellular throughout and are characterized by accumulation of chondroitin sulfate proteoglycans (CSPGs). A comprehensive discussion of the different biological markers that define each lesion type as well as useful clinical markers used to categorize them in patients has been recently reviewed elsewhere (see 22–24). Recently, sub-domains of these lesions have been identified using spatial transcriptomics, further adding complexity to these three main types25. Active lesions are more commonly seen in early RRMS or SPMS, while inactive and chronic active lesions dominate in the non-relapsing phase of SPMS and PPMS19,26,27. Myelin debris engulfed within microglia and macrophages, evident in some active and chronic active lesions, suggests ongoing myelin destruction19,20. Interestingly, despite consistent demyelination OL numbers tend to be preserved in early active lesions, though are increasingly lost in chronic active/inactive lesions28. Whether this ‘dying back’ is the initial step in OL death, or if there are distinct pathologic mechanisms underlying demyelination and OL death remains unclear.
Humoral factors mediating demyelination: immunoglobulins and complement
Humoral factors, including antibodies and complement proteins, are leading candidates in the destruction and clearance of myelin. Antibodies (mainly IgG and to a lesser extent IgM) are deposited in a majority of active, chronic active, and inactive MS lesions21,29–31. While the source of CNS-deposited antibodies is difficult to ascertain, the presence of cerebrospinal fluid (CSF)-restricted oligoclonal bands in most PwMS is evidence of antibody production in or around the CNS likely related to clonally expanded B cells and plasmablasts32,33. Indeed, activated B cells with somatic hypermutations of V(H) genes can be found in the CSF and form follicle-like structures in the subarachnoid space33–35. Antibodies derived from B cells within the subarachnoid space are sufficient to induce demyelination in spinal cord explants36, but despite this, no single dominant autoantigen has been found in MS. A recent study identified a clone of IgG1 antibodies that targeted the proteolipid protein 1 (PLP1), a component of myelin, that could induce MS-like pathology in mice; however, these PLP1-specific antibodies were present in only ~57% of MS patients examined37. A few additional candidate antigens have emerged, with several reports detailing antibody responses at the axo-glial interface to neurofascin and contactin-238–40. One study found that CSF-derived antibodies from MS patients bind to neurofilament light, an axonal protein41, and myelin-reactive “2d2” T cells can recognize neurofilament in experimental autoimmune encephalomyelitis (EAE), an animal model of autoimmune neuroinflammation42. Other reports, however, found similar amounts of neurofilament light chain autoantibodies in control samples or reactivity of CSF-derived antibodies to ubiquitous, non-CNS antigens43,44. More recent studies have confirmed common antigenic motifs seen in Epstein-Barr Virus (EBV) infection and MS, including peptide sequences from the EBV proteins BRRF2 and EBNA45–48. The difficulty in defining the target antigen may be related to both diversity of targets between individuals and epitope spreading, which occurs during the relapsing phase of the disease and broadens the pool of antigen targets.
Despite equipoise around specific targets, the robust deposition of antibodies in MS lesions is suspected to contribute to demyelination. One likely mechanism is the classical complement pathway, an enzymatic chain reaction triggered by antibody deposition that results in formation of opsonins (such as C3b), pro-inflammatory molecules (including the anaphylatoxins C3a and C5a), and the cytolytic C5b-9 complex termed the membrane attack complex (MAC) (Figure 1)49. Large amounts of complement proteins that contribute to the cascade are produced in the liver and released into circulation, though complement is also produced locally by reactive astrocytes and microglia50–53. Several histologic studies have found deposition of antibody alongside activated complement components, including C1q, C3b, and C5b-9, in MS lesions21,29,31,42,54. In addition to activating the classical complement cascade, antibodies are opsonins themselves and affect local immune cell functioning directly through the Fc receptor (FcR)55. Immunoglobulin and complement proteins often colocalize with the myelin component PLP in vesicular structures within macrophages, suggesting PLP is opsonized by these molecules prior to engulfment29. One study provided evidence for the relative importance of complement over FcR signaling in experimental demyelination, finding that MOG IgG–induced demyelination was blocked in C1q−/− but not FcRɣ−/− animals56. The role of complement was complicated by studies finding that complement component C5 deficient mice, which are unable to generate the C5b-9 complex, still had impaired remyelination and enhanced OL death, supporting the idea that earlier complement components such as C3 may be driving pathology57. Despite these findings, it should be noted that in other neurodegenerative disorders, complement inhibition has had mixed results; while most studies report improved outcomes following complement inhibition58, several older studies identified worsening of pathology in Alzheimer’s disease models59,60. The possibility that complement serves a protective function by opsonizing myelin debris and dead OLs should be noted. Without the removal of debris, a persistent inflammatory environment could lead to impaired regeneration.
Figure 1: Complement in inflammatory demyelination.
A) The classical complement cascade. B) Macroscopic deposition of antibody and complement in demyelinating white matter lesions. Antibodies in the perivascular CSF-filled space escape into the brain parenchyma, allowing for deposition of microglial (MG)-secreted C1q and activation of astrocyte (Ast)-secreted C3 into C3a and C3b at the chronic active edge. C) Complement locally influences glia towards reactive phenotypes. Deposition of C3b onto myelin can be recognized by microglial CD11b, resulting in phagocytosis. Stimulation of C3aR with C3a results in increases of HIF-1a as well as alterations in phagocytosis depending on the duration of the stimulation. Made in BioRender.
Cytokine signaling impacts OL survival and function
Cytokines are effector molecules, produced and sensed by infiltrating immune cells and CNS resident cells, that orchestrate immune responses. OLs and OPCs express receptors for many cytokines, and several have direct impacts on survival, proliferation, and differentiation.
TNF is a multi-faceted molecule produced mainly by peripheral immune cells and glia in MS. TNF inhibition is therapeutic in numerous autoimmune conditions, and based on early evidence that it is elevated in MS CSF and CNS tissues, TNF and TNFR antagonists were evaluated as effective therapies in MS. Surprisingly, however, patients treated with the TNF inhibitor lenercept had increased MS exacerbations61. To explain this outcome, subsequent studies explored the nuances of TNF signaling and found opposing effects of signaling through the two TNF receptors, TNFR1 and TNFR2. TNF has two distinct forms, transmembrane (tmTNF) and soluble (solTNF), and TNFR1 binds solTNF or tmTNF, while TNFR2 is activated only by tmTNF62. TNFR1 is closely linked to cell death, harboring an intracellular death domain, and activation of TNFR1 on OLs promotes FAS-mediated apoptosis63,64. TNFR2, in contrast, promotes OL survival and myelination in EAE65. Additionally, TNF signaling alters the inflammatory state of OLs: TNFR1 knockdown in OLs results in a decrease in inflammatory signaling in EAE while knocking down TNFR2 enhances BBB breakdown, microglial activation, and subsequent inflammation65. TNFR2 is also expressed on OPCs, where its activation promotes differentiation, survival, and remyelination65–68. TNFR2−/− animals have impaired progenitor expansion and remyelination after cuprizone66, and TNFR2−/− OPC cultures have impaired survival and differentiation65. Like OLs, OPCs become active participants in neuroinflammation, secreting cytokines and chemokines that attract and activate immune cells. Here too, TNF has opposing effects depending on the receptor activated, with TNFR2 promoting protective and reparative functions of OPCs in vitro69.
Interferon gamma (IFNγ), classically regarded as a master regulator of anti-viral responses, has dose and disease stage dependent effects on myelination. IFNγ induces apoptosis of OLs in vitro70, and overexpression of IFNγ in the mouse brain results in developmental hypomyelination, primary demyelination in early adulthood, and CD8+ T cell-associated OL loss in the aged brain71–73. However, low concentrations of IFNγ actually protect OLs from oxidative stress, and transgenic expression of low levels of IFNγ protects mice against cuprizone-induced demyelination74,75. Lin et al. established the importance of timing in this system, employing a mouse model where IFNγ is inducible downstream of the GFAP promoter, resulting in astrocyte-specific expression of IFNγ. When IFNγ was expressed during the acute phase of EAE, it reduced disease severity, demyelination, and oligodendrocyte loss. However, when IFNγ was expressed during the recovery phase of cuprizone or EAE, remyelination was markedly impaired76. Interestingly, endoplasmic reticulum (ER) stress appears to underlie the impact of IFNγ on OL lineage cells, affecting both demyelination and remyelination76,77. Genetically or pharmacologically increasing the integrated stress response to appropriate levels in OLs increases remyelination in both EAE and cuprizone models78. IFNγ also directly acts on OPCs, reducing proliferation, differentiation, and myelin production in vitro, though its impact on OPCs in vivo remains an area of investigation79,80.
While not directly implicated in OL death like TNF or IFNγ, the impact of numerous other cytokines have been studied on OPCs: IL-17 blocks OPC differentiation and enhances inflammation through induction of NOTCH181; IL-1β and IL-33 both block OPC proliferation and enhance differentiation through activation of the p38-MAPK signaling pathway82–84; IL-4, a driver of allergic-type inflammation, however promotes OPC differentiation through a PPARγ dependent mechanism85; and transforming growth factor beta (TGFβ) regulates OPC differentiation during development and promotes remyelination after experimental demyelination86,87.
The emerging role for inflammatory OLs and OPCs in MS
Beyond their roles in homeostasis and myelination, OLs and OPCs are increasingly appreciated as active participants in neuroinflammation88,89. Major histocompatibility complex type 1 (MHCI) and 2 (MHCII) are cell surface proteins that present self-antigen to cytotoxic T cells or phagocytosed antigen to helper T cells, respectively. Accumulating evidence suggests that IFNγ produces an inflammatory phenotype in OL lineage cells, particularly when combined with IL-17 or TNF. Inflammatory OLs (iOLs) or OPCs (iOPCs) perpetuate tissue inflammation and lose physiologic functions, including remyelination potential88. Early evidence from cultured cells found that OLs upregulate MHCI in response to IFNγ, and subsequent studies confirmed increased MHCI expression in OLs from MS tissue90–93. Interestingly, iOPCs acquire traits usually present only in immune cells, including MHCII expression and the ability to ‘cross-present’ phagocytosed antigens on MHCI94. Recent single cell and single nucleus RNA-seq studies have confirmed OL and OPC upregulation of antigen presentation machinery in EAE and MS95–98. Potentially through this mechanism, iOLs and iOPCs amplify neuroinflammation by activating T cells and secreting cytokines and chemokines89. Indeed, iOLs and iOPCs play a substantial role in neuroinflammation: one study found that deleting ACT1 (a component of the IL-17R signaling cascade) specifically in PDGFRα+ OPCs markedly reduced EAE severity, highlighting the role of OPCs in perpetuating inflammatory IL-17 signaling99. Cytotoxic lymphocytes can also target iOLs and iOPCs for destruction, contributing to demyelination and limiting the pool of cells available to remyelinate94. Further studies are needed, however, to fully clarify their impact on de- and remyelination in MS.
Recent Developments in the Understanding of the Mechanisms of Glial Activation and Glia-Mediated Neurotoxicity in MS.
Microglia and Astrocytes in the setting of neuroinflammation and neurodegeneration
In the context of inflammation-mediated neurodegeneration, glia have diverse functions and can either promote protection and recovery or become deleterious and neurotoxic100. In the healthy brain, microglia and astrocytes perform homeostatic functions that directly or indirectly support to neurons, synapses, oligodendrocytes, and each other. Microglia survey their environment, clearing debris and remodeling neuronal circuitry via pruning of unnecessary synaptic connections101–103. Astrocytes, meanwhile, provide metabolic support to neurons and stabilize synaptic connections104,105. In the setting of inflammation, however, these glial cells can lose their homeostatic functions and enter reactive states characterized broadly by production of inflammatory cytokines, interactions with immune cells, and even direct neurotoxicity, as will be discussed below. In the following sections, we will review what is known about how microglia and astrocytes are activated towards disease-associated states, including the similarities and differences between reactive glia in MS vs other neurodegenerative diseases, and the mechanisms by which these glia mediate neurotoxicity.
Microglia in MS vs other neurodegenerative diseases
While the historic classification of macrophages or microglia into “M1” and “M2” phenotypes is oversimplistic, myeloid cells clearly adopt distinct states in the settings of disease. Activation of microglia can be beneficial, as reparative microglia remove debris and encourage regeneration. For example, a recent study showed how in mice with defects of the PLP1 protein, microglial removal of myelin protected against CD8+ T cell-mediated axonopathy106. However, research in several neurodegenerative diseases has identified subsets of “reactive microglia” as critically involved in their pathogenesis107–109. While there is substantial overlap in microglia phenotype across diseases, resulting in the broad term “Disease-Associated Microglia” (DAM), many fields have generated terms for disease-specific reactive microglia (e.g. “human AD microglia” or HAMs)110,111. MS has recently joined the list of disorders with a specific nomenclature for their DAMs. Sampling of microglia from the chronic active edge of paramagnetic rim lesions (PRLs) in MS has revealed the pathogenic subtype of microglia termed “microglia inflamed in MS” or MIMS which are characterized by elevations in C1Q, FTH1, CD68, and additional iron-regulatory genes112. Single nucleus transcriptomics have identified that microglia have distinct genetic signatures in MS compared to both secondary demyelination and healthy controls113. In particular, while both secondary demyelination and MS microglia have decreases in classic homeostatic markers like P2RY12 and CX3CR1, MS microglia exhibit increases in FTH1, SIPA1L1, and ACSL1 whereas secondary demyelination show higher levels of MYO1E and APOE113. Therefore, it is important to consider that while most neurodegenerative or demyelinating disorders have similar microglial phenotypes, there are key distinctions that could have important implications for the development of microglia-targeting therapies.
Mechanisms of microglial activation in MS
Blood-brain barrier (BBB) disruption is a prominent feature of acute MS lesions. Gadolinium enhancing lesions have been shown to correlate with perivenular cuffs of immune cells which release matrix metalloproteases that allow them to traverse the endothelium. Demyelinating lesions radiating outward from the post-capillary venules are rapidly becoming a specific marker of MS and can be detected using magnetic resonance imaging-based acquisitions such as T2*114,115. T and B cells that encounter cognate antigen are reactivated in the CNS where they release cytokines that mediate microglial activation. Incubation of microglia with IFNγ, TNF, and IL-1β results in activation of mTOR signaling and subsequent production of cytotoxic nitric oxide116. A recent paper identified additional cytokines that may selectively trigger pro-inflammatory polarization in the setting of MS: knockdown of IFIT3, largely produced by migrating macrophages, reduced microglial ability to polarize towards this reactive phenotype in the spinal cords of EAE mice possibly via attenuation of STAT1 and NF-kB signaling117.
Exposure of microglia to bloodborne humoral elements also results in activation of inflammatory transcriptional profiles. In EAE, focused ultrasound-induced BBB disruption resulted in focal alterations in the activation state of microglia despite no change in amount of cellular infiltrate118. In particular, fibrinogen (a key member of the clotting cascade) directly signals the CD11b receptor on myeloid lineage cells and induces oxidative stress pathways, redox regulation, and type 1 IFN gene families as well as clustering of microglia in the perivascular space119–121. Fibrinogen-stimulated microglia recreate the oxidative stress signature of microglia in EAE, suggesting that exposure of microglia to fibrinogen from blood may be a key signal in their polarization towards a reactive state.
As discussed previously, the complement cascade has been studied for its contribution to demyelination in the setting of inflammation (Figure 1) and has recently been shown to be associated with MS disease severity122. The complement components C3a and C5a promote inflammation and leukocyte invasion123,124. Signaling of C3a onto the C3a receptor (C3aR) on microglia results in upregulation of hypoxia-inducible factor 1a (HIF-1a) leading to depletion of ATP stores, metabolic impairment, and aberrant microglial response in a model of AD125. C3aR signaling also has biphasic effects on phagocytosis: short term administration promotes phagocytosis whereas longer term (24 hour) stimulation stifles phagocytosis126. The other members of the early complement cascade are gaining more attention as well. The ability of non-anaphylatoxin members of the complement cascade to directly trigger pro-inflammatory transcriptional changes in microglia has recently come to the forefront121. Stimulation of primary microglia with the opsonizing form of C3 (iC3b) results in oxidative stress responses, phospholipid metabolic processes, and reorganization of the extracellular matrix121 with specific upregulation of additional complement genes such as C1QA. This may be due to direct signaling through the complement receptor 3 (CR3; aka CD11b), which is expressed by microglia. A recent study showed that siRNA-mediated knockdown of CR3 attenuates activation of microglia in the setting of chronic inflammation caused by rotenone127. This resulted in subsequent reduction in the production of C1q, TNF, and IL-1α, a cocktail of cytokines termed “TIC” that is known to induce neurotoxic reactive astrocytes in multiple neurodegenerative diseases51. C3 therefore has multiple mechanisms by which it can signal through microglia to promote reactive and phagocytic phenotypes.
Astrocytes in MS vs other neurodegenerative diseases
Recent studies have identified a broad heterogeneity in astrocyte subtypes in different disorders. While there are some overlapping differentially expressed genes (DEGs) in spinal cord injury, LPS-induced neuroinflammation, and EAE, a recent study only found 2.6% of all astrocyte reactivity DEGs were present in all three128. A RiboTag approach to examining regional differences in astrocyte transcript profiles in EAE showed distinct pathways especially in spinal cord vs brain derived astrocytes129. Clearly many subtypes of astrocytes exist but are still referred to under the broad multi-disorder banner of “reactive astrocytes”130,131.
In MS, single cell profiling demonstrates that astrocytes decrease their expression of the nuclear factor erythroid 2-related factor 2 (NRF2), a master transcriptional regulator of the endogenous anti-oxidant response, while simultaneously upregulating MAFG, a small Maf (sMAF) transcription factor involved in promoting pro-inflammatory transcriptional profiles132. In contrast, in AD, MAFG has been suggested to not be upregulated whereas a different sMAF protein (MAFF) has been suggested to be the most responsive sMAF protein to AD pathology133–135, though the cellular source of MAFF in AD has not been determined yet. While these sMAF proteins can both form heterodimers with NRF2 to initiate antioxidant and anti-inflammatory transcription, they can form homodimers which lack a transcriptional activation domain and subsequently repress transcriptional programs136,137. Upregulation of sMAFs such as MAFG in MS and MAFF in AD without concurrent upregulation of NRF2 therefore represents a mechanism by which astrocytes can repress their antioxidant and anti-inflammatory profiles.
Phosphorylation of the ER stress sensor IRE1a following stimulation of astrocytes with pro-inflammatory cytokines such as IL-1β and TNF results in full translation of the transcription factor XBP1, which has been shown to drive a subset of astrocytes towards pathogenicity in MS138–140. This pathogenicity was in part due to the increased ability of XBP1+ astrocytes to recruit peripheral monocytes to the CNS and promote microglial activation. IRE1a-XBP1 signaling in astrocytes is stimulated by the accumulation of unfolded proteins and activation of the unfolded protein response.
Recent work on MS patient induced pluripotent stem cell-derived astrocytes demonstrated increased processing and presentation of antigen as well as genes related to EBV response compared to healthy controls, even in the absence of inflammatory stimuli141. These data suggest an intrinsic factor in MS astrocytes that predisposes them towards reactive states which may contribute to development of pathology, a finding potentially corroborated by the discovery of epigenetic signatures in EAE that prime astrocytes towards reactivity142. The upregulation of EBV-related genes in MS astrocytes strongly supports the recent evidence that highlights a role for EBV infection in predisposing individuals to developing MS, a topic which has been extensively reviewed elsewhere (see 46,143,144).
Microglia-astrocyte crosstalk contributes to astrocyte activation in MS
Astrocytes are central figures in the signaling milieu during neuroinflammation, modulating peripheral immune infiltration and chronic tissue inflammation (reviewed in 145). Independent of peripheral immune signaling, microglia and astrocytes can also engage in crosstalk that results in development of reactive astrocytes. It has been well described that following NLRP3 inflammasome action on Caspase-1, microglia can stimulate reactive astrocytes via the secretion of the TIC cocktail of cytokines51,146. Upon stimulation with TIC, astrocytes take on a phenotype characterized by increased complement production and neurotoxicity. Additional signaling between microglia and astrocytes has become evident. The MIMS discussed above have been shown to interact closely with reactive astrocytes in chronic active MS lesions largely via microglial C1Q and astrocytic C3112. Intriguingly, in a cell culture model of AD, Guttikonda et al. showed that microglia require the ability to produce C3 in order to initiate reciprocal signaling with astrocytes which subsequently produced excess C3147. It is possible that microglial production of C3 is therefore also vital to activate neurotoxic astrocytes in MS. The ability to investigate astrocyte signaling interactions has improved with a recently developed technique termed RABID-seq in which a viral-based barcoding system allows transmission of the virus between interacting cells so that cell-cell interactions can be tracked followed by single-cell (sc)RNA-sequencing. This technique demonstrated that microglia and astrocytes interact via the axon guidance molecules Sema4D-PlexinB2 and EphrinB3-EphB3 in EAE, and that inhibition of EphB3 suppresses reactive astrocyte formation and ameliorates EAE148.
Another example of microglial-astrocyte crosstalk mediating glial activation involves the master transcriptional regulator of the inflammatory response, NF-κB, Its activation has distinct consequences in different cell types, with NF-κB activation appearing to protect OLs during EAE149,150 but also potentially enhancing pathologically reactive astrocytes. Reactive astrocytes show increased NF-κB activation in both MS and EAE151. A study by Ponath et al. found a known MS-risk variant, rs7665090G, located nearby the NFKB1 locus which increased NF-κB signaling and gene expression (e.g., CXCL10, C3) in human astrocyte cell cultures as well as in astrocytes within MS lesions, suggesting a potential astrocyte-mediated causal link to pathogenesis152. Of note, a recent study utilized a microfluidic platform to analyze cell-cell interactions and identified that astrocytic NF-κB activation (experimentally induced by stimulation with LPS) can be suppressed by signaling from microglial-derived amphiregulin (an epidermal growth factor), suggesting a potential glial-crosstalk system that regulates astrocyte NF-κB153. Intriguingly, they found that microglial expression of amphiregulin could be induced by astrocyte-derived IL-33, completing an anti-inflammatory signaling loop between astrocytes and microglia. It may be possible that this glial interaction is altered in MS, although this has not yet been investigated. With recent observations that MS risk genes are also expressed in microglia and that 18 of the known MS risk gene variants are related to the NF-κB and AP-1 signaling pathway it is plausible that excessive glial responsiveness or failed NF-κB regulation may account for chronic CNS inflammation154,155.
Mechanisms of microglial and astrocyte neurotoxicity
Following pathological activation, microglia and astrocytes contribute to neurotoxicity in MS and other neurodegenerative diseases. The proposed mechanisms by which they mediate this injury are many and varied (Figure 2). One of these is glial generation of and interactions with the complement system. In the past, investigation of the role of the complement system in MS was largely focused on direct cytotoxic injury through MAC57,156. However, more recent attention has been paid towards early factors, including C1Q and C3. Astrocytes are the primary producers of C3 in the CNS, and numerous studies have highlighted how astrocytic C3 is associated with worse pathology in MS and EAE50,157–159. The mechanism by which C3 mediates neurodegeneration is still unclear, however. C1Q and C3 have both been identified as key contributors to the excessive elimination of synapses in AD and MS via opsonization and subsequent phagocytosis160–164. Glia-mediated synapse loss has also been linked to the Signal regulatory protein α (SIRPα) in microglia, via its interactions with the “don’t eat me” signal CD47 (Figure 2)165. The direct link between synapse loss and neurodegeneration is unclear, however. While it may be possible that excessive elimination of synapses on otherwise healthy neurons is a driving step towards cell death, it conversely is possible that already injured neurons may signal for phagocytes to remove synapses that may contribute to excessive excitatory signaling that could cause oxidative stress, and recent discussion has questioned whether aberrant synaptic “stripping” is involved in disease pathogenesis166.
Figure 2: Proposed Mechanisms of Glial Neurotoxicity in PMS.
A) Normal healthy CNS environment, with CD47 (a “Don’t Eat Me” signal) signaling through microglial Sirpα to halt phagocytosis of synapses. Homeostatic astrocytes support neuronal metabolism and ensure that neurons have sufficient resources to thrive. B) The setting of neuroinflammation in the CNS. Reactive astrocytes upregulate glycolysis and translation, leading to diversion of resources away from neurons and towards glia, resulting in a state of virtual hypoxia. Astrocytes may directly mediate neurodegeneration through secretion of Long Chain Fatty Acids (LCFAs) and TNF among other cytokines, while microglial Galectin-3 enhances the neurotoxicity of oxidized phosphatidylcholine (OxPC). Meanwhile, stressed neurons downregulate CD47 concurrent with increases in local complement deposition, resulting in phagocytosis of synapses which may lead to neurodegeneration. Made in BioRender.
The search for a glia-derived directly neurotoxic factor has resulted in a few candidates. A variety of other microglia- and astrocyte-derived molecules have been previously reviewed for their potential to cause neurotoxicity167,168. One of the more well-studied mediators of neurodegeneration is astrocyte-secreted TNF which can interact with TNF receptors on neurons to initiate Fas-mediated apoptosis169–172. A few additional candidates have recently emerged; for example, Guttenplan et al reported that long-chain saturated lipids produced by astrocytes mediated neurotoxicity both in vitro and in vivo173. Additionally, microglial Galectin-3, a lectin associated with pro-inflammatory conditions, exacerbates the neurotoxicity generated by oxidized phosphatidylcholines, lipids elevated in EAE and MS174,175. In numerous neurodegenerative models, the enzyme SARM1 has been shown to play crucial roles in axonal injury with subsequent neuronal loss176–178. Astrocytes upregulate the SARM1 in EAE, which results in reductions of glial-derived neurotrophic factor (GDNF) and subsequent demyelination and neuroinflammation179. A recent study suggested that SARM1 may also mediate its pathological effects on axons by increasing NF-κB signaling following reduction of the NF-κB inhibitor IGFBP2180. Deletion of SARM1 only blocks axonal degeneration in the early phases of EAE, however, and at late timepoints, SARM1 knockout mice were indistinguishable from wild types, indicating that SARM1’s contribution to axonal degeneration may vary over the course of CNS inflammation181. Meanwhile, in the neurodegenerative diseases amyotrophic lateral sclerosis and frontotemporal dementia, astrocytes secrete elevated levels of inorganic polyphosphate, and neutralizing this inorganic polyphosphate alleviates neurotoxicity182. Neither of these molecules have been investigated in MS yet, however, and warrant further research.
Another postulated mechanism of neurodegeneration in the setting of MS is metabolic stress and virtual hypoxia. Virtual hypoxia is a state in which neurons are deficient in metabolic resources like glucose, oxygen, and energy, potentially due to mitochondrial damage, oxidative injury, or the excessive demand of resources by reactive glia183. Several studies have demonstrated shifts in amino acid metabolism184 and increases in oxidative stress in PwMS185, implicating metabolic disruption in disease pathogenesis. Microglia exposed to inflammatory stimuli such as LPS or NO shift their metabolic programming towards glycolysis, and re-focusing them towards oxidative phosphorylation via administration of dimethyl malonate generates anti-inflammatory transcriptional programs186. In astrocytes, a shift towards increased glycolysis results in elevated inflammation187,188. This suggests that when glia acquire highly energy-demanding phenotypes, there is a concomitant increase in inflammation and neuronal stress, potentially due to a paucity of available resources for neurons. The protective effects on myelin of promoting the integrated stress response in oligodendrocytes has been well described79,189–191 although the subsequent effects on neurodegeneration have not been as well explored. Diversion of resources away from metabolically-demanding reactive glia and towards vulnerable neurons may be transiently neuroprotective as well as result in an anti-inflammatory environment that might promote downstream regeneration.
Therapeutic approaches targeting non-relapsing progressive disease and remyelination: Lessons learned and future directions
Limited Efficacy of Remyelinating and Neuroprotective Drugs in Clinical Trials in People Living with MS
Disease modifying therapeutic strategies for the treatment of PwMS are currently limited to immunomodulatory drugs that inhibit acute inflammation in the CNS. Current high-efficacy disease-modifying therapies (DMTs) for MS limit the activation or CNS infiltration of the adaptive immune system, thereby greatly reducing relapse rates192. However, these therapies have little to no impact on myelin repair or halting neurodegeneration in progressive MS. Over the last two decades, several potential pro-remyelination and neuroprotective therapies have been developed based on the concept that OPCs are abundant and proliferative cells in the CNS and can be driven to differentiate into myelinating OLs upon withdrawal of PDGFRα, induction with triiodothyronine, and a number of other pharmacological compounds193. Indeed, exciting preclinical data support roles for anti-muscarinic agents (e.g. clemastine, benztropine), anti-histamine (GSK239512), RXR agonism (bexarotene, clobetasol), LINGO antagonists (opicinumab), and cholesterol biogenesis (emopamil binding protein (EBP) inhibitors) in vitro and in animal models leading to early phase trials of these putative remyelinating agents in PwMS194. Despite these preclinical successes, however, several clinical trials of potential remyelinating and neuroprotective therapies in PwMS have yielded negative or underwhelming results, which we will review below.
LINGO-1 is a transmembrane cell surface glycoprotein that regulates OPC differentiation and proliferation as well as axonal growth195. Genetic deletion of LINGO-1, knockdown of LINGO-1, or treatment with anti-LINGO-1 antibodies results in increased axonal myelination and neuronal survival in cell culture and animal models, including cuprizone and MOG-induced EAE196–200. The anti-LINGO-1 antibody opicinumab initially showed improvement in visual evoked potential (VEP) testing in people with optic neuritis in the per-protocol analysis of the Phase II RENEW trial201. Despite these encouraging results, opicinumab failed to reach its primary end points in both the SYNERGY and AFFINITY phase II trials in PwMS202. After AFFINITY, clinical development of opicinumab was discontinued.
Another potential small molecule remyelination therapy, clemastine, was identified in 2014 using a micropillar array high-throughput screening platform203. Similarly to opicinumab, clemastine showed promise in several animal models of demyelination204,205. The Phase II ReBUILD trial suggested that clemastine treatment in PwMS with chronic demyelinating optic neuropathy might promote endogenous remyelination of the optic nerve as measured by reduced P100 latency delay of 1.7ms/eye on VEP testing206. The small magnitude of this effect is of unclear clinical significance. A phase IIa clinical trial of RXR-agonist bexarotene in patients with RRMS failed to reach its primary efficacy outcome of improved lesional magnetization transfer ratio (MTR, a correlate of myelin density) and furthermore demonstrated poor tolerability with significantly increased adverse events in the bexarotene-treated group including central hypothyroidism (100%), hypertriglyceridemia (92%), rash (50%), and neutropenia (38%)207. In a post-hoc exploratory analysis of this trial, significant treatment effects were seen at the whole lesion level in gray matter but not white matter lesions, and voxel-level analyses detected significant treatment effects in white matter lesion voxels with the lowest baseline MTR, suggesting these measures may be more sensitive to myelin repair208. In contrast to bexarotene, a phase II study of GSK239512, a well-tolerated brain penetrant histamine H3 receptor blocker, in PwMS showed a small but statistically significant effect on remyelination as measured by changes in lesional MTR209.
In addition to pro-remyelination therapies, several direct neuroprotective strategies have been tested in early phase clinical trials in both RRMS and PMS with the majority failing to reach their primary endpoints. The Phase IIb MS-SMART trial assessed the efficacy of three axonal-targeted neuroprotective drugs in a cohort of 393 patients with SPMS: amiloride that limits Na+ and Ca2+-dependent neuroaxonal injury, riluzole that reduces glutamate excitotoxicity, and fluoxetine which has pleiotropic effects on energy metabolism210. The primary outcome of brain volume change on MRI was not met in any arms despite an adequately powered trial, suggesting that targeting these pathological mechanisms of axonal injury was insufficient to ameliorate neuroaxonal loss in SPMS over the course of the trial. Multiple other putative neuroprotective therapies have shown negative or minimally positive results in clinical trials of PwMS (reviewed in 211). In sum, recent years have yielded ineffective or minimally efficacious clinical trials with respect to remyelination or neuroprotection in PwMS. Identifying reasons why these compounds and trials have had limited success is vital to improving future progress in MS care.
Barriers to Remyelination and Neuroprotection
Many of the remyelinating strategies tested in MS clinical trials target OPC differentiation and/or proliferation directly. However, the presence of non-differentiating OPCs within chronic MS lesions, albeit at lower densities than normal white matter, suggests that OPC proliferation and differentiation may be extrinsically inhibited in these lesions28,96,212–214. Over the last decade, researchers have identified several extrinsic factors that can interfere with OPC biology and may prevent therapies from effectively targeting OPCs and promoting remyelination. Similarly, many of the neuroprotective strategies for PwMS directly target neurons as opposed to reactive glia and extrinsic factors that contribute to a pro-neurodegenerative environment. As outlined above, compartmentalized chronic inflammation within the CNS (including reactive glia, immune cells, and pro-inflammatory cytokines) can interfere with effective endogenous and exogenous remyelination and neuroprotection signals. Additional extrinsic factors including changes in the extracellular matrix (e.g. chondroitin sulfate proteoglycans/CSPGs), leaky BBB, blood products including fibrinogen, axon receptivity, myelin debris, and metabolic stress may also contribute to this anti-remyelination and pro-neurodegenerative environment211.
A study using human induced pluripotent stem cell-derived OLs (hiOLs) showed that blockage of OL differentiation with activated PBMC-supernatants, an effect mediated at least in part through IFNγ, could not be restored by treatment with the OPC differentiation-promoting drugs clemastine, benztropine, or miconazole. However, OL differentiation was partially restored in this model by pre-treatment of the activated-PBMCs with the anti-inflammatory drug teriflunomide215. Other studies have similarly showed several pro-myelinating compounds including clemastine, benztropine, miconazole, and clobetasol were unable to overcome fibrinogen’s or CSPGs’ extrinsic inhibition of OPC differentiation216,217. These studies demonstrate the capability of MS-relevant extrinsic factors to inhibit the therapeutic efficacy of multiple remyelination therapeutic compounds that directly target OPCs.
The identification of effective novel therapeutic compounds for remyelination and neuroprotection is limited in part by the current animal models used to pre-clinically assess such compounds. Many demyelinating models such as the cuprizone chemical-demyelinating model do not accurately reflect the microenvironment and chronic inflammatory milieu associated with demyelinated plaques and progressive disease in PwMS, and inflammatory models such as EAE are accompanied by extensive axonal pathology with few demyelinated axons amenable to remyelination. Development of more relevant models is paramount for the discovery of effective therapies.
Overcoming Barriers to Remyelination and Neuroprotection
Including Extrinsic Inhibitory Factors in Pre-Clinical Screening Models
One strategy to develop drugs that overcome the barrier posed by the inhibitory microenvironment of MS lesions is to incorporate identified inhibitory signals into remyelination and neuroprotection screening assays. Several groups have developed such models that begin to address this issue. Petersen et al. added fibrinogen to primary rat OPC differentiation media prior to screening for myelin-promoting compounds in order to mimic one key component of the inhibitory lesion environment. In this screen, they found that 7 previously-identified pro-OPC-differentiation compounds failed to work in the presence of fibrinogen; however, the BMP type I receptor inhibitor DMH1 was identified as a novel molecule that could restore OPC differentiation to mature oligodendrocytes despite the presence of fibrinogen216.
In addition to in vitro screens, extrinsic inhibitory factors can be incorporated animal models of remyelination and neurodegeneration. For example, the toxin cuprizone induces demyelination, and effective remyelination occurs when it is stopped. Several models have been developed in which cuprizone is combined with CNS inflammation, which may more closely resemble an MS lesion, such as (i) adoptive transfer of myelin reactive Th17 cells into previously cuprizone-fed mice218,219, (ii) immune boost of complete Freund’s adjuvant and pertussis toxin without exogenous myelin peptides in cuprizone-fed mice220, and (iii) cuprizone-fed mice with ectopically-expressed inducible IFNγ production in astrocytes (GFAP/tTA mice x TRE/IFNγ)77. While improved, these models may still not adequately model the chronic gliosis, fibrinogen, or deposition of CSPGs that inhibit remyelination at later stages of lesion evolution. In the IFNγ-cuprizone model, which allows for assessment of OPC differentiation and remyelination in an IFNγ-high environment, prolonging the integrated stress response with the compound Sephin1 increased the number of remyelinating oligodendrocytes and remyelinated axons. However, the thickness of axonal myelin only returned to near pre-lesion levels when Sephin1 treatment was combined with the OPC pro-differentiation compound bazedoxifene78. These in vitro and in vivo studies underscore the importance of screening for therapies in a more relevant environment and the potential benefit of combinatorial approaches to promote remyelination and neuroprotection in an inflammatory microenvironment.
Compartmentalized CNS Inflammation
Using the novel methodologies described above, new compounds have been identified that can promote remyelination in the setting of individual extrinsic inhibitory factors. However, a strategy that may work in one inhibitory environment (e.g. high levels of IFNγ) may not work in another (e.g. excessive fibrinogen). Many, but not all, of the identified factors that promote the perilesional microenvironment associated with inhibition of remyelination and ongoing neurodegeneration are downstream of persistently reactive and pro-inflammatory glia. Thus, a two-pronged approach of targeting OPCs in addition to reactive glia may more effectively facilitate myelin repair in PwMS. A similar approach targeting intrinsic pro-survival pathways in neurons while inhibiting pro-cell-death extrinsic signals from reactive glia may be needed for an effective neuroprotective approach in chronic inflammatory diseases.
Current high-efficacy DMTs such as anti-CD20 and anti-alpha4 integrin monoclonal antibodies are highly protective against clinical relapses and new acute inflammatory lesions in RRMS, but do not target the compartmentalized CNS inflammation and reactive glia observed in PMS. Interestingly, multiple other DMTs that are not as effective against clinical relapses may potentially target CNS compartmentalized inflammation. Dimethyl fumarate (DMF) has well-described inhibitory effects on NF-κB pathway activation, an innate immune pathway highly upregulated in disease-associated microglia and astrocytes in MS112. DMF decreases susceptibility in PRLs 2.3x faster than glatiramer acetate, suggesting DMF may improve the pathology of chronic active lesions221, a correlate of neurodegeneration and disease progression in MS. Identifying therapeutic targets that can effectively dampen the smoldering inflammation and innate immune activation within the CNS of PwMS is a critical unmet need.
Emerging Strategies for Limiting Neurodegeneration and Promoting Remyelination in MS
Targeting CNS Compartmentalized Inflammation
Given the increasing appreciation of CNS-compartmentalized inflammation in PwMS, several potential strategies to target CNS disease-associated microglia, astrocytes, macrophages, and B-cells are under investigation. Bruton tyrosine kinase (BTK) is an intracellular signaling protein that regulates activation, differentiation, and survival of B-cells, microglia, and possibly astrocytes222, and the efficacy of several BTK-inhibitors (BTKIs) on MS relapses and progressive disease are currently being investigated in Phase III clinical trials. In the evolutionRMS 1 (NCT04338022) and evolutionRMS 2 (NCT04338061) trials, the only BTKI Phase III trials in PwMS with reported results, the BTKI evobrutinib failed to show superiority to oral teriflunomide in reduction of annualized relapse rate, potentially partly due to an unexpectedly low relapse rate in the teriflunomide arm of the study. While trials of other BTKIs in RMS are ongoing, these results suggest that BTKI monotherapy may not be highly efficacious against acute MS relapses. However, the inhibition of microglial and astrocytic BTK may still have benefits in targeting glial inflammation and thus may still represent a possibly beneficial drug for patients with non-relapsing PMS. Multiple BTKIs are in clinical trials for non-relapsing SPMS (NCT04411641) and PPMS (NCT04458051, NCT04544449).
Targeting the complement pathway in MS is another potentially viable therapeutic option that may modulate CNS-compartmentalized glial inflammation. Of note, a C3 inhibitor pegcetacoplan was recently FDA approved for treatment of macular degeneration, and the C5-blocking antibody eculizamab is FDA approved for neuromyelitis optica, highlighting the potential for translating complement-targeting drugs into human patients. Inhibition of signaling between the opsonizing form of C3 and its receptor CR2 with a pair of CR2-conjugated inhibitors ameliorated both acute and chronic disability in EAE223. The same group found that inhibition of the alternative complement pathway with a monoclonal antibody against complement factor B (mAb 1379) reduced chronic demyelination without affecting the acute phase of the disease, implying it may have promise in progressive MS224. More recently, treatment of EAE mice with a monoclonal antibody inhibitor of C1q (ANX-M1.21) reduced the concentration of MIMS in the hippocampi of EAE mice at chronic timepoints, highlighting the role that complement might play in fostering inflammatory environments112. In addition, an AAV-Crry was recently shown to rescue visual function and synaptic pathology in EAE162. However, despite these preclinical successes, no clinical trial has attempted to investigate complement inhibitors in PMS.
Several other therapeutic strategies that target glial activation are under investigation in clinical and pre-clinical studies. Inhibitors of other tyrosine kinases involved in innate immune activation include mastinib which inhibits the c-Kit pathway225. A randomized, double-blind Phase III clinical trial showed that low-dose, but peculiarly not high-dose, mastinib treatment was associated with minimally less disease progression as measured by change EDSS from baseline in patients with PPMS and non-relapsing SPMS226. Astroglial toll-like receptor (TLR) signaling, including through TLR4, also have central roles in innate immune activation and have been shown to contribute to neuroinflammation and MS pathogenesis227,228. A phase II trial in PMS has shown an association between treatment with the anti-TLR4, phosphodiesterase type 5 (PDE5)-inhibiting, and macrophage migration inhibitor factor (MIF)-inhibiting compound ibudilast and slower progression of brain atrophy in progressive MS229.
The growing evidence of the role of glial activation and CNS compartmentalized-inflammation in the chronic neurodegeneration observed in progressive MS and the early, albeit minimal, positive signals of the strategies above have led to increased pre-clinical identification and testing of several other potential therapeutic targets to regulate the innate immune system and glial activation in MS. Evaluation of these targets should include determining whether they can decrease the expression or release of inhibitory factors as described above from reactive glia and in demyelination models. It should further be determined whether there are synergistic effects when these glial-targeting therapies are used in combination with remyelination and neuroprotective therapies that target OPCs and neurons, respectively.
Targeting Metabolism
Additional therapeutic strategies related to modulating metabolic pathways have shown some promise in immunomodulation, neuroprotection, and remyelination. α-lipoic acid, a naturally occurring antioxidant that has immunomodulatory effects on both the adaptive and innate immune systems230, is associated with slower progression of brain atrophy in a phase II trial in PMS231. Statins have been shown in several models to have anti-inflammatory and neuroprotective effects, and the MS-STAT1 Phase II trial showed that high-dose simvastatin was well tolerated and reduced the annualized rate of whole brain atrophy by 43% in patients with SPMS232. A multi-center Phase III trial of high-dose simvastatin verses placebo in SPMS (MS-STAT2) is currently ongoing with results expected in 2025 (NCT03387670). Circulating bile acid metabolites have been shown to be lower in PwMS compared to healthy controls and this alteration in bile acid metabolism may relate to alterations in the gut microbiome233,234. The endogenous bile acid, tauroursodeoxycholic acid (TUDCA), has been shown to prevent neurotoxic polarization of astrocytes and pro-inflammatory polarization of microglia in vitro and reduce severity of EAE233. Current evidence points to a potentially protective effect of TUDCA in multiple neurodegenerative diseases, including amyotrophic lateral sclerosis, Alzheimer’s disease, Huntington’s disease, and Parkinson’s disease235,236. A recent observational cohort study demonstrated that higher primary bile acid levels at baseline predicted slower whole brain and retinal layer atrophy in PMS and that TUDCA supplementation was safe, increased serum levels of multiple bile acids, decreased circulating central memory CD4+ and Th1/17 cells, and increased CD4+ naive cells compared to placebo237. As expected, TUDCA supplementation was also associated with alterations in the composition and function of gut microbiota. Additional pre-clinical screens have identified multiple endogenous metabolites that promote OPC differentiation at physiological-relevant levels including taurine, an aminosulfonic acid, which was able to increase the efficacy of drug-induced OPC differentiation with benztropine and miconazole238. These studies highlight the growing appreciation of metabolism and immunometabolism and the potential of several metabolic strategies to modulate glial-activation, neuronal survival, and remyelination in PwMS.
Disinhibiting CNS Repair: Targeting Inhibitory Factors
In addition to inflammation and reactive glia, the identification of several specific extrinsic factors that may inhibit remyelination has led to pursuit of therapeutic approaches directly targeting some of these factors. Anti-fibrin immunotherapy, which can enter CNS and bind parenchymal fibrin without interfering with clotting, inhibited amyloid-associated and autoimmune demyelination-driven neurotoxicity in animal models239. The defibrinogenating agent ancrod has also been shown to enhance remyelination in the lysolecithin focal demyelination model239. Similarly, anti-CSPG compounds that competitively inhibit the synthesis of CSPGs by astrocytes were able to reduce CSPG expression and promoted remyelination in vitro and in vivo217,240. These studies demonstrate the potential clinical benefit of targeting detrimental extrinsic factors, such as fibrinogen and CSPGs, to ameliorate neurotoxicity and facilitate CNS repair. Antagonists of these and other inhibitory signals of OPC differentiation may themselves promote remyelination and may also increase the efficacy of pro-remyelination therapies when used in combination.
Targeting Novel Cell Death Pathways
Another emerging avenue for neuroprotection and oligoprotection in MS is targeting recently-identified novel cell death pathways. Necroptosis is a form of necrotic cell death where inflammatory stimuli, including TLR4 activation and TNFR1 signaling among others, activate RIPK1, leading to formation of the necroptosome241. Ferroptosis is another form of necrotic cell death driven by iron-dependent phospholipid peroxidation and regulated by multiple metabolic and signaling pathways242. Upon extensive DNA damage, poly-ADP ribose (PAR) polymerase-1 (PARP-1) can initiate the form of cell death termed parthanatos that is ultimately executed by macrophage migration inhibitory factor (MIF) nuclease activity243.
Necroptosis, ferroptosis, and parthanatos have been implicated in neurodegeneration in PwMS244–246, as well as in several other neurodegenerative diseases241,247–249. Moreover, inhibition of necroptosis, ferroptosis, and parthanatos with RIPK1 inhibitors, ferrostatin-1 analogs, and PARP-1 or MIF nuclease inhibitors like PAANIB-1 respectively reduce neuronal loss in neurodegenerative diseases244–246. Necroptosis, ferroptosis, and parthanatos signaling have all been shown to contribute to the generation of neurotoxic microglia and astrocytes244,250–252. Additionally, glial ferroptosis and necroptosis have been shown to reduce OPC differentiation and OL myelinating capacity244,253. Thus, inhibiting these cell death pathways may both directly prevent OL and neuronal cell death as well as mitigate disease-state associated oligotoxic and neurotoxic glia.
Conclusions
The need for new therapies that address the chronic neurodegenerative aspect of MS is growing. Despite an increasing understanding of the mechanisms by which resident glia of the CNS participate in these chronic inflammatory processes that drive this neurodegeneration, there is a paucity of novel therapeutics to address the progressive phase of MS. As we have discussed, there is a dire need for animal models that faithfully recapitulate progressive disease and remyelination in the context of inflammation. Additionally, the goal of effectively remyelinating axons in the setting of MS requires targeting both OPCs directly as well suppressing the inflammatory microenvironment presented by reactive astrocytes and microglia. Finally, in order to adequately treat PwMS, we need to specifically measure outcomes that correspond to CNS inflammation, progressive neurodegeneration, and remyelination in future clinical trials. Together, evaluation of MS as a complex, multi-faceted disease may afford future clinical trials more success.
Figure 3: Pharmacologic efforts to promote myelination and neuroprotection.
A) An oligodendroglial cell showing the targets for drugs that have been shown to promote oligodendroglial differentiation, maturation, and/or myelination. Several identified inhibitory factors of remyelination are also depicted with flathead arrows directed to the drugs that these inhibitors been shown to target. Notably, DMH1 is not inhibited by fibrinogen and Sephin1 upregulates the ISR even in the presence of IFNγ. B) Selection of key drugs and molecules that may help downregulate neurotoxic and oligotoxic reactive glia. Also displayed are pharmacologic inhibitors of the non-apoptotic cell death pathways necroptosis, ferroptosis, and parthanatos. Made in BioRender.
Funding
TG is supported by NIH T32 GM136577. SPG is supported by NMSS and ABF FAN-2106-37832. AJG is supported by NMSS and ABF FAN-2007-36944. PAC is supported by NIH R01NS041435, DOD W81XWH1910622, and NMSS RG-1907-34756
Footnotes
Declaration of Interests
TG, SPG, and AJG declare no competing interests. PAC has received consulting fees from Eli Lilly and Novartis and is a PI on a grant to Johns Hopkins University from Genentech.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Bibliography
- 1.Kuhlmann T, Moccia M, Coetzee T, Cohen JA, Correale J, Graves J, Marrie RA, Montalban X, Yong VW, Thompson AJ, et al. (2023). Multiple sclerosis progression: time for a new mechanism-driven framework. Lancet Neurol 22, 78–88. 10.1016/S1474-4422(22)00289-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Fransen NL, de Jong BA, Hess K, Kuhlmann T, Vincenten MCJ, Hamann J, Huitinga I, and Smolders J (2021). Absence of B Cells in Brainstem and White Matter Lesions Associates With Less Severe Disease and Absence of Oligoclonal Bands in MS. Neurol Neuroimmunol Neuroinflamm 8. 10.1212/NXI.0000000000000955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Fransen NL, Hsiao CC, van der Poel M, Engelenburg HJ, Verdaasdonk K, Vincenten MCJ, Remmerswaal EBM, Kuhlmann T, Mason MRJ, Hamann J, et al. (2020). Tissue-resident memory T cells invade the brain parenchyma in multiple sclerosis white matter lesions. Brain 143, 1714–1730. 10.1093/brain/awaa117. [DOI] [PubMed] [Google Scholar]
- 4.Machado-Santos J, Saji E, Troscher AR, Paunovic M, Liblau R, Gabriely G, Bien CG, Bauer J, and Lassmann H (2018). The compartmentalized inflammatory response in the multiple sclerosis brain is composed of tissue-resident CD8+ T lymphocytes and B cells. Brain 141, 2066–2082. 10.1093/brain/awy151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Moccia M, Haider L, Eshaghi A, van de Pavert SHP, Brescia Morra V, Patel A, Wheeler-Kingshott CAM, Barkhof F, and Ciccarelli O (2022). B Cells in the CNS at Postmortem Are Associated With Worse Outcome and Cell Types in Multiple Sclerosis. Neurol Neuroimmunol Neuroinflamm 9. 10.1212/NXI.0000000000001108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.van Nierop GP, van Luijn MM, Michels SS, Melief MJ, Janssen M, Langerak AW, Ouwendijk WJD, Hintzen RQ, and Verjans G (2017). Phenotypic and functional characterization of T cells in white matter lesions of multiple sclerosis patients. Acta Neuropathol 134, 383–401. 10.1007/s00401-017-1744-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Tonietto M, Poirion E, Lazzarotto A, Ricigliano V, Papeix C, Bottlaender M, Bodini B, and Stankoff B (2023). Periventricular remyelination failure in multiple sclerosis: a substrate for neurodegeneration. Brain 146, 182–194. 10.1093/brain/awac334. [DOI] [PubMed] [Google Scholar]
- 8.Mey GM, Mahajan KR, and DeSilva TM (2023). Neurodegeneration in multiple sclerosis. WIREs Mech Dis 15, e1583. 10.1002/wsbm.1583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kang SH, Fukaya M, Yang JK, Rothstein JD, and Bergles DE (2010). NG2+ CNS glial progenitors remain committed to the oligodendrocyte lineage in postnatal life and following neurodegeneration. Neuron 68, 668–681. 10.1016/j.neuron.2010.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Wheeler NA, and Fuss B (2016). Extracellular cues influencing oligodendrocyte differentiation and (re)myelination. Exp Neurol 283, 512–530. 10.1016/j.expneurol.2016.03.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Grinspan J (2002). Cells and signaling in oligodendrocyte development. J Neuropathol Exp Neurol 61, 297–306. 10.1093/jnen/61.4.297. [DOI] [PubMed] [Google Scholar]
- 12.Buchanan J, Elabbady L, Collman F, Jorstad NL, Bakken TE, Ott C, Glatzer J, Bleckert AA, Bodor AL, Brittain D, et al. (2022). Oligodendrocyte precursor cells ingest axons in the mouse neocortex. Proc Natl Acad Sci U S A 119, e2202580119. 10.1073/pnas.2202580119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Lin SC, and Bergles DE (2002). Physiological characteristics of NG2-expressing glial cells. J Neurocytol 31, 537–549. 10.1023/a:1025799816285. [DOI] [PubMed] [Google Scholar]
- 14.Gallo V, and Armstrong RC (2008). Myelin repair strategies: a cellular view. Curr Opin Neurol 21, 278–283. 10.1097/WCO.0b013e3282fd1875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Orthmann-Murphy J, Call CL, Molina-Castro GC, Hsieh YC, Rasband MN, Calabresi PA, and Bergles DE (2020). Remyelination alters the pattern of myelin in the cerebral cortex. Elife 9. 10.7554/eLife.56621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lucchinetti C, Bruck W, Parisi J, Scheithauer B, Rodriguez M, and Lassmann H (1999). A quantitative analysis of oligodendrocytes in multiple sclerosis lesions. A study of 113 cases. Brain 122 ( Pt 12), 2279–2295. 10.1093/brain/122.12.2279. [DOI] [PubMed] [Google Scholar]
- 17.Yeung MSY, Djelloul M, Steiner E, Bernard S, Salehpour M, Possnert G, Brundin L, and Frisen J (2019). Dynamics of oligodendrocyte generation in multiple sclerosis. Nature 566, 538–542. 10.1038/s41586-018-0842-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Reich DS, Lucchinetti CF, and Calabresi PA (2018). Multiple Sclerosis. N Engl J Med 378, 169–180. 10.1056/NEJMra1401483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kuhlmann T, Ludwin S, Prat A, Antel J, Bruck W, and Lassmann H (2017). An updated histological classification system for multiple sclerosis lesions. Acta Neuropathol 133, 13–24. 10.1007/s00401-016-1653-y. [DOI] [PubMed] [Google Scholar]
- 20.Popescu BF, and Lucchinetti CF (2012). Pathology of demyelinating diseases. Annu Rev Pathol 7, 185–217. 10.1146/annurev-pathol-011811-132443. [DOI] [PubMed] [Google Scholar]
- 21.Lucchinetti C, Bruck W, Parisi J, Scheithauer B, Rodriguez M, and Lassmann H (2000). Heterogeneity of multiple sclerosis lesions: implications for the pathogenesis of demyelination. Ann Neurol 47, 707–717. . [DOI] [PubMed] [Google Scholar]
- 22.Akesson J, Hojjati S, Hellberg S, Raffetseder J, Khademi M, Rynkowski R, Kockum I, Altafini C, Lubovac-Pilav Z, Mellergard J, et al. (2023). Proteomics reveal biomarkers for diagnosis, disease activity and long-term disability outcomes in multiple sclerosis. Nat Commun 14, 6903. 10.1038/s41467-023-42682-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Frisch T, Elkjaer ML, Reynolds R, Michel TM, Kacprowski T, Burton M, Kruse TA, Thomassen M, Baumbach J, and Illes Z (2020). Multiple Sclerosis Atlas: A Molecular Map of Brain Lesion Stages in Progressive Multiple Sclerosis. Netw Syst Med 3, 122–129. 10.1089/nsm.2020.0006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kolb H, Al-Louzi O, Beck ES, Sati P, Absinta M, and Reich DS (2022). From pathology to MRI and back: Clinically relevant biomarkers of multiple sclerosis lesions. Neuroimage Clin 36, 103194. 10.1016/j.nicl.2022.103194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kukanja P, Langseth CM, Rubio Rodriguez-Kirby LA, Agirre E, Zheng C, Raman A, Yokota C, Avenel C, Tiklova K, Guerreiro-Cacais AO, et al. (2024). Cellular architecture of evolving neuroinflammatory lesions and multiple sclerosis pathology. Cell 187, 1990–2009 e1919. 10.1016/j.cell.2024.02.030. [DOI] [PubMed] [Google Scholar]
- 26.Frischer JM, Weigand SD, Guo Y, Kale N, Parisi JE, Pirko I, Mandrekar J, Bramow S, Metz I, Bruck W, et al. (2015). Clinical and pathological insights into the dynamic nature of the white matter multiple sclerosis plaque. Ann Neurol 78, 710–721. 10.1002/ana.24497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Luchetti S, Fransen NL, van Eden CG, Ramaglia V, Mason M, and Huitinga I (2018). Progressive multiple sclerosis patients show substantial lesion activity that correlates with clinical disease severity and sex: a retrospective autopsy cohort analysis. Acta Neuropathol 135, 511–528. 10.1007/s00401-018-1818-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Hess K, Starost L, Kieran NW, Thomas C, Vincenten MCJ, Antel J, Martino G, Huitinga I, Healy L, and Kuhlmann T (2020). Lesion stage-dependent causes for impaired remyelination in MS. Acta Neuropathol 140, 359–375. 10.1007/s00401-020-02189-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Munoz U, Sebal C, Escudero E, Esiri M, Tzartos J, Sloan C, and Sadaba MC (2022). Main Role of Antibodies in Demyelination and Axonal Damage in Multiple Sclerosis. Cell Mol Neurobiol 42, 1809–1827. 10.1007/s10571-021-01059-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Sadaba MC, Tzartos J, Paino C, Garcia-Villanueva M, Alvarez-Cermeno JC, Villar LM, and Esiri MM (2012). Axonal and oligodendrocyte-localized IgM and IgG deposits in MS lesions. J Neuroimmunol 247, 86–94. 10.1016/j.jneuroim.2012.03.020. [DOI] [PubMed] [Google Scholar]
- 31.Breij EC, Brink BP, Veerhuis R, van den Berg C, Vloet R, Yan R, Dijkstra CD, van der Valk P, and Bo L (2008). Homogeneity of active demyelinating lesions in established multiple sclerosis. Ann Neurol 63, 16–25. 10.1002/ana.21311. [DOI] [PubMed] [Google Scholar]
- 32.Beltran E, Gerdes LA, Hansen J, Flierl-Hecht A, Krebs S, Blum H, Ertl-Wagner B, Barkhof F, Kumpfel T, Hohlfeld R, and Dornmair K (2019). Early adaptive immune activation detected in monozygotic twins with prodromal multiple sclerosis. J Clin Invest 129, 4758–4768. 10.1172/JCI128475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Qin Y, Duquette P, Zhang Y, Talbot P, Poole R, and Antel J (1998). Clonal expansion and somatic hypermutation of V(H) genes of B cells from cerebrospinal fluid in multiple sclerosis. J Clin Invest 102, 1045–1050. 10.1172/JCI3568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Haugen M, Frederiksen JL, and Degn M (2014). B cell follicle-like structures in multiple sclerosis-with focus on the role of B cell activating factor. J Neuroimmunol 273, 1–7. 10.1016/j.jneuroim.2014.05.010. [DOI] [PubMed] [Google Scholar]
- 35.Franciotta D, Salvetti M, Lolli F, Serafini B, and Aloisi F (2008). B cells and multiple sclerosis. Lancet Neurol 7, 852–858. 10.1016/S1474-4422(08)70192-3. [DOI] [PubMed] [Google Scholar]
- 36.Blauth K, Soltys J, Matschulat A, Reiter CR, Ritchie A, Baird NL, Bennett JL, and Owens GP (2015). Antibodies produced by clonally expanded plasma cells in multiple sclerosis cerebrospinal fluid cause demyelination of spinal cord explants. Acta Neuropathol 130, 765–781. 10.1007/s00401-015-1500-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Owens GP, Fellin TJ, Matschulat A, Salas V, Schaller KL, Given KS, Ritchie AM, Navarro A, Blauth K, Hughes EG, et al. (2023). Pathogenic myelin-specific antibodies in multiple sclerosis target conformational proteolipid protein 1-anchored membrane domains. J Clin Invest 133. 10.1172/JCI162731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Derfuss T, Linington C, Hohlfeld R, and Meinl E (2010). Axo-glial antigens as targets in multiple sclerosis: implications for axonal and grey matter injury. J Mol Med (Berl) 88, 753–761. 10.1007/s00109-010-0632-3. [DOI] [PubMed] [Google Scholar]
- 39.Derfuss T, Parikh K, Velhin S, Braun M, Mathey E, Krumbholz M, Kumpfel T, Moldenhauer A, Rader C, Sonderegger P, et al. (2009). Contactin-2/TAG-1-directed autoimmunity is identified in multiple sclerosis patients and mediates gray matter pathology in animals. Proc Natl Acad Sci U S A 106, 8302–8307. 10.1073/pnas.0901496106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Mathey EK, Derfuss T, Storch MK, Williams KR, Hales K, Woolley DR, Al-Hayani A, Davies SN, Rasband MN, Olsson T, et al. (2007). Neurofascin as a novel target for autoantibody-mediated axonal injury. J Exp Med 204, 2363–2372. 10.1084/jem.20071053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Silber E, Semra YK, Gregson NA, and Sharief MK (2002). Patients with progressive multiple sclerosis have elevated antibodies to neurofilament subunit. Neurology 58, 1372–1381. 10.1212/wnl.58.9.1372. [DOI] [PubMed] [Google Scholar]
- 42.Krishnamoorthy G, Saxena A, Mars LT, Domingues HS, Mentele R, Ben-Nun A, Lassmann H, Dornmair K, Kurschus FC, Liblau RS, and Wekerle H (2009). Myelin-specific T cells also recognize neuronal autoantigen in a transgenic mouse model of multiple sclerosis. Nat Med 15, 626–632. 10.1038/nm.1975. [DOI] [PubMed] [Google Scholar]
- 43.Willis SN, Stathopoulos P, Chastre A, Compton SD, Hafler DA, and O’Connor KC (2015). Investigating the Antigen Specificity of Multiple Sclerosis Central Nervous System-Derived Immunoglobulins. Front Immunol 6, 600. 10.3389/fimmu.2015.00600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Brandle SM, Obermeier B, Senel M, Bruder J, Mentele R, Khademi M, Olsson T, Tumani H, Kristoferitsch W, Lottspeich F, et al. (2016). Distinct oligoclonal band antibodies in multiple sclerosis recognize ubiquitous self-proteins. Proc Natl Acad Sci U S A 113, 7864–7869. 10.1073/pnas.1522730113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Cepok S, Zhou D, Srivastava R, Nessler S, Stei S, Bussow K, Sommer N, and Hemmer B (2005). Identification of Epstein-Barr virus proteins as putative targets of the immune response in multiple sclerosis. J Clin Invest 115, 1352–1360. 10.1172/JCI23661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lanz TV, Brewer RC, Ho PP, Moon JS, Jude KM, Fernandez D, Fernandes RA, Gomez AM, Nadj GS, Bartley CM, et al. (2022). Clonally expanded B cells in multiple sclerosis bind EBV EBNA1 and GlialCAM. Nature 603, 321–327. 10.1038/s41586-022-04432-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Roshan HM, Valadan R, Hosseini SA, and Ajami A (2023). Specific antibodies to EBNA1 epitopes as risk factors for multiple sclerosis, a systematic review. J Neuroimmunol 383, 578202. 10.1016/j.jneuroim.2023.578202. [DOI] [PubMed] [Google Scholar]
- 48.Zamecnik CR, Sowa GM, Abdelhak A, Dandekar R, Bair RD, Wade KJ, Bartley CM, Tubati A, Gomez R, Fouassier C, et al. (2023). A Predictive Autoantibody Signature in Multiple Sclerosis. medRxiv. 10.1101/2023.05.01.23288943. [DOI] [Google Scholar]
- 49.Mastellos DC, Hajishengallis G, and Lambris JD (2023). A guide to complement biology, pathology and therapeutic opportunity. Nat Rev Immunol. 10.1038/s41577-023-00926-1. [DOI] [PubMed] [Google Scholar]
- 50.Gharagozloo M, Smith MD, Jin J, Garton T, Taylor M, Chao A, Meyers K, Kornberg MD, Zack DJ, Ohayon J, et al. (2021). Complement component 3 from astrocytes mediates retinal ganglion cell loss during neuroinflammation. Acta Neuropathol 142, 899–915. 10.1007/s00401-021-02366-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, Bennett ML, Munch AE, Chung WS, Peterson TC, et al. (2017). Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541, 481–487. 10.1038/nature21029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Hong S, Beja-Glasser VF, Nfonoyim BM, Frouin A, Li S, Ramakrishnan S, Merry KM, Shi Q, Rosenthal A, Barres BA, et al. (2016). Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science 352, 712–716. 10.1126/science.aad8373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Zhang LY, Pan J, Mamtilahun M, Zhu Y, Wang L, Venkatesh A, Shi R, Tu X, Jin K, Wang Y, et al. (2020). Microglia exacerbate white matter injury via complement C3/C3aR pathway after hypoperfusion. Theranostics 10, 74–90. 10.7150/thno.35841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ingram G, Loveless S, Howell OW, Hakobyan S, Dancey B, Harris CL, Robertson NP, Neal JW, and Morgan BP (2014). Complement activation in multiple sclerosis plaques: an immunohistochemical analysis. Acta Neuropathol Commun 2, 53. 10.1186/2051-5960-2-53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Nimmerjahn F, and Ravetch JV (2008). Fcgamma receptors as regulators of immune responses. Nat Rev Immunol 8, 34–47. 10.1038/nri2206. [DOI] [PubMed] [Google Scholar]
- 56.Urich E, Gutcher I, Prinz M, and Becher B (2006). Autoantibody-mediated demyelination depends on complement activation but not activatory Fc-receptors. Proc Natl Acad Sci U S A 103, 18697–18702. 10.1073/pnas.0607283103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Rus H, Cudrici C, and Niculescu F (2005). C5b-9 complement complex in autoimmune demyelination and multiple sclerosis: dual role in neuroinflammation and neuroprotection. Ann Med 37, 97–104. 10.1080/07853890510007278. [DOI] [PubMed] [Google Scholar]
- 58.Dalakas MC, Alexopoulos H, and Spaeth PJ (2020). Complement in neurological disorders and emerging complement-targeted therapeutics. Nat Rev Neurol 16, 601–617. 10.1038/s41582-020-0400-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Maier M, Peng Y, Jiang L, Seabrook TJ, Carroll MC, and Lemere CA (2008). Complement C3 deficiency leads to accelerated amyloid beta plaque deposition and neurodegeneration and modulation of the microglia/macrophage phenotype in amyloid precursor protein transgenic mice. J Neurosci 28, 6333–6341. 10.1523/JNEUROSCI.0829-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Wyss-Coray T, Yan F, Lin AH, Lambris JD, Alexander JJ, Quigg RJ, and Masliah E (2002). Prominent neurodegeneration and increased plaque formation in complement-inhibited Alzheimer’s mice. Proc Natl Acad Sci U S A 99, 10837–10842. 10.1073/pnas.162350199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.TNF neutralization in MS: results of a randomized, placebo-controlled multicenter study. The Lenercept Multiple Sclerosis Study Group and The University of British Columbia MS/MRI Analysis Group. (1999). Neurology 53, 457–465. [PubMed] [Google Scholar]
- 62.Probert L (2015). TNF and its receptors in the CNS: The essential, the desirable and the deleterious effects. Neuroscience 302, 2–22. 10.1016/j.neuroscience.2015.06.038. [DOI] [PubMed] [Google Scholar]
- 63.Hovelmeyer N, Hao Z, Kranidioti K, Kassiotis G, Buch T, Frommer F, von Hoch L, Kramer D, Minichiello L, Kollias G, et al. (2005). Apoptosis of oligodendrocytes via Fas and TNF-R1 is a key event in the induction of experimental autoimmune encephalomyelitis. J Immunol 175, 5875–5884. 10.4049/jimmunol.175.9.5875. [DOI] [PubMed] [Google Scholar]
- 64.Wajant H, and Siegmund D (2019). TNFR1 and TNFR2 in the Control of the Life and Death Balance of Macrophages. Front Cell Dev Biol 7, 91. 10.3389/fcell.2019.00091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Madsen PM, Motti D, Karmally S, Szymkowski DE, Lambertsen KL, Bethea JR, and Brambilla R (2016). Oligodendroglial TNFR2 Mediates Membrane TNF-Dependent Repair in Experimental Autoimmune Encephalomyelitis by Promoting Oligodendrocyte Differentiation and Remyelination. J Neurosci 36, 5128–5143. 10.1523/JNEUROSCI.0211-16.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Arnett HA, Mason J, Marino M, Suzuki K, Matsushima GK, and Ting JP (2001). TNF alpha promotes proliferation of oligodendrocyte progenitors and remyelination. Nat Neurosci 4, 1116–1122. 10.1038/nn738. [DOI] [PubMed] [Google Scholar]
- 67.Fischer R, Padutsch T, Bracchi-Ricard V, Murphy KL, Martinez GF, Delguercio N, Elmer N, Sendetski M, Diem R, Eisel ULM, et al. (2019). Exogenous activation of tumor necrosis factor receptor 2 promotes recovery from sensory and motor disease in a model of multiple sclerosis. Brain Behav Immun 81, 247–259. 10.1016/j.bbi.2019.06.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Maier O, Fischer R, Agresti C, and Pfizenmaier K (2013). TNF receptor 2 protects oligodendrocyte progenitor cells against oxidative stress. Biochem Biophys Res Commun 440, 336–341. 10.1016/j.bbrc.2013.09.083. [DOI] [PubMed] [Google Scholar]
- 69.Desu HL, Illiano P, Choi JS, Ascona MC, Gao H, Lee JK, and Brambilla R (2021). TNFR2 Signaling Regulates the Immunomodulatory Function of Oligodendrocyte Precursor Cells. Cells 10. 10.3390/cells10071785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Peferoen L, Kipp M, van der Valk P, van Noort JM, and Amor S (2014). Oligodendrocyte-microglia cross-talk in the central nervous system. Immunology 141, 302–313. 10.1111/imm.12163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Horwitz MS, Evans CF, McGavern DB, Rodriguez M, and Oldstone MBA (1997). Primary demyelination in transgenic mice expressing interferon-gamma. Nat Med 3, 1037–1041. 10.1038/nm0997-1037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.LaFerla FM, Sugarman MC, Lane TE, and Leissring MA (2000). Regional hypomyelination and dysplasia in transgenic mice with astrocyte-directed expression of interferon-gamma. J Mol Neurosci 15, 45–59. 10.1385/JMN:15:1:45. [DOI] [PubMed] [Google Scholar]
- 73.Kaya T, Mattugini N, Liu L, Ji H, Cantuti-Castelvetri L, Wu J, Schifferer M, Groh J, Martini R, Besson-Girard S, et al. (2022). CD8(+) T cells induce interferon-responsive oligodendrocytes and microglia in white matter aging. Nat Neurosci 25, 1446–1457. 10.1038/s41593-022-01183-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Gao X, Gillig TA, Ye P, D’Ercole AJ, Matsushima GK, and Popko B (2000). Interferon-gamma protects against cuprizone-induced demyelination. Mol Cell Neurosci 16, 338–349. 10.1006/mcne.2000.0883. [DOI] [PubMed] [Google Scholar]
- 75.Balabanov R, Strand K, Goswami R, McMahon E, Begolka W, Miller SD, and Popko B (2007). Interferon-gamma-oligodendrocyte interactions in the regulation of experimental autoimmune encephalomyelitis. J Neurosci 27, 2013–2024. 10.1523/JNEUROSCI.4689-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Lin W, Kemper A, Dupree JL, Harding HP, Ron D, and Popko B (2006). Interferon-gamma inhibits central nervous system remyelination through a process modulated by endoplasmic reticulum stress. Brain 129, 1306–1318. 10.1093/brain/awl044. [DOI] [PubMed] [Google Scholar]
- 77.Lin W, Harding HP, Ron D, and Popko B (2005). Endoplasmic reticulum stress modulates the response of myelinating oligodendrocytes to the immune cytokine interferon-gamma. J Cell Biol 169, 603–612. 10.1083/jcb.200502086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Chen Y, Kunjamma RB, Weiner M, Chan JR, and Popko B (2021). Prolonging the integrated stress response enhances CNS remyelination in an inflammatory environment. Elife 10. 10.7554/eLife.65469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Chen Y, Quan S, Patil V, Kunjamma RB, Tokars HM, Leisten ED, Joy G, Wills S, Chan JR, Wong YC, and Popko B (2023). Insights into the mechanism of oligodendrocyte protection and remyelination enhancement by the integrated stress response. Glia 71, 2180–2195. 10.1002/glia.24386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Saraswat D, Welliver RR, Ravichandar R, Tripathi A, Polanco JJ, Broome J, Hurley E, Dutta R, Feltri ML, and Sim FJ (2021). Heparanome-Mediated Rescue of Oligodendrocyte Progenitor Quiescence following Inflammatory Demyelination. J Neurosci 41, 2245–2263. 10.1523/JNEUROSCI.0580-20.2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Wang C, Zhang CJ, Martin BN, Bulek K, Kang Z, Zhao J, Bian G, Carman JA, Gao J, Dongre A, et al. (2017). IL-17 induced NOTCH1 activation in oligodendrocyte progenitor cells enhances proliferation and inflammatory gene expression. Nat Commun 8, 15508. 10.1038/ncomms15508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ljunggren-Rose A, Natarajan C, Matta P, Pandey A, Upender I, and Sriram S (2020). Anacardic acid induces IL-33 and promotes remyelination in CNS. Proc Natl Acad Sci U S A 117, 21527–21535. 10.1073/pnas.2006566117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Natarajan C, Yao SY, and Sriram S (2016). TLR3 Agonist Poly-IC Induces IL-33 and Promotes Myelin Repair. PLoS One 11, e0152163. 10.1371/journal.pone.0152163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Vela JM, Molina-Holgado E, Arevalo-Martin A, Almazan G, and Guaza C (2002). Interleukin-1 regulates proliferation and differentiation of oligodendrocyte progenitor cells. Mol Cell Neurosci 20, 489–502. 10.1006/mcne.2002.1127. [DOI] [PubMed] [Google Scholar]
- 85.Zhang Q, Zhu W, Xu F, Dai X, Shi L, Cai W, Mu H, Hitchens TK, Foley LM, Liu X, et al. (2019). The interleukin-4/PPARgamma signaling axis promotes oligodendrocyte differentiation and remyelination after brain injury. PLoS Biol 17, e3000330. 10.1371/journal.pbio.3000330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Hamaguchi M, Muramatsu R, Fujimura H, Mochizuki H, Kataoka H, and Yamashita T (2019). Circulating transforming growth factor-beta1 facilitates remyelination in the adult central nervous system. Elife 8. 10.7554/eLife.41869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Palazuelos J, Klingener M, and Aguirre A (2014). TGFbeta signaling regulates the timing of CNS myelination by modulating oligodendrocyte progenitor cell cycle exit through SMAD3/4/FoxO1/Sp1. J Neurosci 34, 7917–7930. 10.1523/JNEUROSCI.0363-14.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Kirby L, and Castelo-Branco G (2021). Crossing boundaries: Interplay between the immune system and oligodendrocyte lineage cells. Semin Cell Dev Biol 116, 45–52. 10.1016/j.semcdb.2020.10.013. [DOI] [PubMed] [Google Scholar]
- 89.Zeis T, Enz L, and Schaeren-Wiemers N (2016). The immunomodulatory oligodendrocyte. Brain Res 1641, 139–148. 10.1016/j.brainres.2015.09.021. [DOI] [PubMed] [Google Scholar]
- 90.Turnley AM, Miller JF, and Bartlett PF (1991). Regulation of MHC molecules on MBP positive oligodendrocytes in mice by IFN-gamma and TNF-alpha. Neurosci Lett 123, 45–48. 10.1016/0304-3940(91)90154-l. [DOI] [PubMed] [Google Scholar]
- 91.Suzumura A, Silberberg DH, and Lisak RP (1986). The expression of MHC antigens on oligodendrocytes: induction of polymorphic H-2 expression by lymphokines. J Neuroimmunol 11, 179–190. 10.1016/0165-5728(86)90002-0. [DOI] [PubMed] [Google Scholar]
- 92.Wong GH, Bartlett PF, Clark-Lewis I, Battye F, and Schrader JW (1984). Inducible expression of H-2 and Ia antigens on brain cells. Nature 310, 688–691. 10.1038/310688a0. [DOI] [PubMed] [Google Scholar]
- 93.Hoftberger R, Aboul-Enein F, Brueck W, Lucchinetti C, Rodriguez M, Schmidbauer M, Jellinger K, and Lassmann H (2004). Expression of major histocompatibility complex class I molecules on the different cell types in multiple sclerosis lesions. Brain Pathol 14, 43–50. 10.1111/j.1750-3639.2004.tb00496.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kirby L, Jin J, Cardona JG, Smith MD, Martin KA, Wang J, Strasburger H, Herbst L, Alexis M, Karnell J, et al. (2019). Oligodendrocyte precursor cells present antigen and are cytotoxic targets in inflammatory demyelination. Nat Commun 10, 3887. 10.1038/s41467-019-11638-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Falcao AM, van Bruggen D, Marques S, Meijer M, Jakel S, Agirre E, Samudyata, Floriddia EM., Vanichkina DP., Ffrench-Constant C., et al. (2018). Disease-specific oligodendrocyte lineage cells arise in multiple sclerosis. Nat Med 24, 1837–1844. 10.1038/s41591-018-0236-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Jakel S, Agirre E, Mendanha Falcao A, van Bruggen D, Lee KW, Knuesel I, Malhotra D, Ffrench-Constant C, Williams A, and Castelo-Branco G (2019). Altered human oligodendrocyte heterogeneity in multiple sclerosis. Nature 566, 543–547. 10.1038/s41586-019-0903-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Kenigsbuch M, Bost P, Halevi S, Chang Y, Chen S, Ma Q, Hajbi R, Schwikowski B, Bodenmiller B, Fu H, et al. (2022). A shared disease-associated oligodendrocyte signature among multiple CNS pathologies. Nat Neurosci 25, 876–886. 10.1038/s41593-022-01104-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Schirmer L, Velmeshev D, Holmqvist S, Kaufmann M, Werneburg S, Jung D, Vistnes S, Stockley JH, Young A, Steindel M, et al. (2019). Neuronal vulnerability and multilineage diversity in multiple sclerosis. Nature 573, 75–82. 10.1038/s41586-019-1404-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Kang Z, Wang C, Zepp J, Wu L, Sun K, Zhao J, Chandrasekharan U, DiCorleto PE, Trapp BD, Ransohoff RM, and Li X (2013). Act1 mediates IL-17-induced EAE pathogenesis selectively in NG2+ glial cells. Nat Neurosci 16, 1401–1408. 10.1038/nn.3505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Schirmer L, Schafer DP, Bartels T, Rowitch DH, and Calabresi PA (2021). Diversity and Function of Glial Cell Types in Multiple Sclerosis. Trends Immunol 42, 228–247. 10.1016/j.it.2021.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Prinz M, Jung S, and Priller J (2019). Microglia Biology: One Century of Evolving Concepts. Cell 179, 292–311. 10.1016/j.cell.2019.08.053. [DOI] [PubMed] [Google Scholar]
- 102.Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mardinly AR, Yamasaki R, Ransohoff RM, Greenberg ME, Barres BA, and Stevens B (2012). Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 74, 691–705. 10.1016/j.neuron.2012.03.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS, Nouri N, Micheva KD, Mehalow AK, Huberman AD, Stafford B, et al. (2007). The classical complement cascade mediates CNS synapse elimination. Cell 131, 1164–1178. 10.1016/j.cell.2007.10.036. [DOI] [PubMed] [Google Scholar]
- 104.Allen NJ, and Eroglu C (2017). Cell Biology of Astrocyte-Synapse Interactions. Neuron 96, 697–708. 10.1016/j.neuron.2017.09.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Sofroniew MV, and Vinters HV (2010). Astrocytes: biology and pathology. Acta Neuropathol 119, 7–35. 10.1007/s00401-009-0619-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Groh J, Abdelwahab T, Kattimani Y, Horner M, Loserth S, Gudi V, Adalbert R, Imdahl F, Saliba AE, Coleman M, et al. (2023). Microglia-mediated demyelination protects against CD8(+) T cell-driven axon degeneration in mice carrying PLP defects. Nat Commun 14, 6911. 10.1038/s41467-023-42570-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Hirbec H, Rassendren F, and Audinat E (2019). Microglia Reactivity: Heterogeneous Pathological Phenotypes. Methods Mol Biol 2034, 41–55. 10.1007/978-1-4939-9658-2_4. [DOI] [PubMed] [Google Scholar]
- 108.Paolicelli RC, Sierra A, Stevens B, Tremblay ME, Aguzzi A, Ajami B, Amit I, Audinat E, Bechmann I, Bennett M, et al. (2022). Microglia states and nomenclature: A field at its crossroads. Neuron 110, 3458–3483. 10.1016/j.neuron.2022.10.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Wang J, He W, and Zhang J (2023). A richer and more diverse future for microglia phenotypes. Heliyon 9, e14713. 10.1016/j.heliyon.2023.e14713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Kenkhuis B, Somarakis A, de Haan L, Dzyubachyk O, ME IJ, de Miranda N, Lelieveldt BPF, Dijkstra J, van Roon-Mom WMC, Hollt T, and van der Weerd L (2021). Iron loading is a prominent feature of activated microglia in Alzheimer’s disease patients. Acta Neuropathol Commun 9, 27. 10.1186/s40478-021-01126-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Srinivasan K, Friedman BA, Etxeberria A, Huntley MA, van der Brug MP, Foreman O, Paw JS, Modrusan Z, Beach TG, Serrano GE, and Hansen DV (2020). Alzheimer’s Patient Microglia Exhibit Enhanced Aging and Unique Transcriptional Activation. Cell Rep 31, 107843. 10.1016/j.celrep.2020.107843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Absinta M, Maric D, Gharagozloo M, Garton T, Smith MD, Jin J, Fitzgerald KC, Song A, Liu P, Lin JP, et al. (2021). A lymphocyte-microglia-astrocyte axis in chronic active multiple sclerosis. Nature 597, 709–714. 10.1038/s41586-021-03892-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Lam M, Lee D, Kosater I, Khairallah A, Taga M, Zhang Y, Fujita M, Nag S, Bennett DA, De Jager P, and Menon V (2023). Human disease-specific cell signatures in non-lesional tissue in Multiple Sclerosis detected by single-cell and spatial transcriptomics. bioRxiv. 10.1101/2023.12.20.572491. [DOI] [Google Scholar]
- 114.Cagol A, Cortese R, Barakovic M, Schaedelin S, Ruberte E, Absinta M, Barkhof F, Calabrese M, Castellaro M, Ciccarelli O, et al. (2023). Diagnostic Performance of Cortical Lesions and the Central Vein Sign in Multiple Sclerosis. JAMA Neurol. 10.1001/jamaneurol.2023.4737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Chaaban L, Safwan N, Moussa H, El-Sammak S, Khoury SJ, and Hannoun S (2022). Central vein sign: A putative diagnostic marker for multiple sclerosis. Acta Neurol Scand 145, 279–287. 10.1111/ane.13553. [DOI] [PubMed] [Google Scholar]
- 116.Dello Russo C, Lisi L, Tringali G, and Navarra P (2009). Involvement of mTOR kinase in cytokine-dependent microglial activation and cell proliferation. Biochem Pharmacol 78, 1242–1251. 10.1016/j.bcp.2009.06.097. [DOI] [PubMed] [Google Scholar]
- 117.Sun R, Wang YF, and Yang X (2024). Knockdown of IFIT3 ameliorates multiple sclerosis via selectively regulating M1 polarization of microglia in an experimental autoimmune encephalomyelitis model. Int Immunopharmacol 128, 111501. 10.1016/j.intimp.2024.111501. [DOI] [PubMed] [Google Scholar]
- 118.Schregel K, Baufeld C, Palotai M, Meroni R, Fiorina P, Wuerfel J, Sinkus R, Zhang YZ, McDannold N, White PJ, and Guttmann CRG (2021). Targeted Blood Brain Barrier Opening With Focused Ultrasound Induces Focal Macrophage/Microglial Activation in Experimental Autoimmune Encephalomyelitis. Front Neurosci 15, 665722. 10.3389/fnins.2021.665722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Davalos D, Ryu JK, Merlini M, Baeten KM, Le Moan N, Petersen MA, Deerinck TJ, Smirnoff DS, Bedard C, Hakozaki H, et al. (2012). Fibrinogen-induced perivascular microglial clustering is required for the development of axonal damage in neuroinflammation. Nat Commun 3, 1227. 10.1038/ncomms2230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Kim S, Sharma C, Jung UJ, and Kim SR (2023). Pathophysiological Role of Microglial Activation Induced by Blood-Borne Proteins in Alzheimer’s Disease. Biomedicines 11. 10.3390/biomedicines11051383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Mendiola AS, Yan Z, Dixit K, Johnson JR, Bouhaddou M, Meyer-Franke A, Shin MG, Yong Y, Agrawal A, MacDonald E, et al. (2023). Defining blood-induced microglia functions in neurodegeneration through multiomic profiling. Nat Immunol 24, 1173–1187. 10.1038/s41590-023-01522-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Oechtering J, Stein K, Schaedelin SA, Maceski AM, Orleth A, Meier S, Willemse E, Qureshi F, Heijnen I, Regeniter A, et al. (2024). Complement Activation Is Associated With Disease Severity in Multiple Sclerosis. Neurol Neuroimmunol Neuroinflamm 11, e200212. 10.1212/NXI.0000000000200212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Ghosh M, and Rana S (2023). The anaphylatoxin C5a: Structure, function, signaling, physiology, disease, and therapeutics. Int Immunopharmacol 118, 110081. 10.1016/j.intimp.2023.110081. [DOI] [PubMed] [Google Scholar]
- 124.Yadav MK, Maharana J, Yadav R, Saha S, Sarma P, Soni C, Singh V, Saha S, Ganguly M, Li XX, et al. (2023). Molecular basis of anaphylatoxin binding, activation, and signaling bias at complement receptors. Cell 186, 4956–4973 e4921. 10.1016/j.cell.2023.09.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Gedam M, Comerota MM, Propson NE, Chen T, Jin F, Wang MC, and Zheng H (2023). Complement C3aR depletion reverses HIF-1alpha-induced metabolic impairment and enhances microglial response to Abeta pathology. J Clin Invest 133. 10.1172/JCI167501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Lian H, Litvinchuk A, Chiang AC, Aithmitti N, Jankowsky JL, and Zheng H (2016). Astrocyte-Microglia Cross Talk through Complement Activation Modulates Amyloid Pathology in Mouse Models of Alzheimer’s Disease. J Neurosci 36, 577–589. 10.1523/JNEUROSCI.2117-15.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Wang Q, Ruan Z, Jing L, Guo Z, Zhang X, Liu J, Tian L, Sun W, Song S, Hong JS, et al. (2023). Complement receptor 3-mediated neurotoxic glial activation contributes to rotenone-induced cognitive decline in mice. Ecotoxicol Environ Saf 266, 115550. 10.1016/j.ecoenv.2023.115550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Burda JE, O’Shea TM, Ao Y, Suresh KB, Wang S, Bernstein AM, Chandra A, Deverasetty S, Kawaguchi R, Kim JH, et al. (2022). Divergent transcriptional regulation of astrocyte reactivity across disorders. Nature 606, 557–564. 10.1038/s41586-022-04739-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Itoh N, Itoh Y, Tassoni A, Ren E, Kaito M, Ohno A, Ao Y, Farkhondeh V, Johnsonbaugh H, Burda J, et al. (2018). Cell-specific and region-specific transcriptomics in the multiple sclerosis model: Focus on astrocytes. Proc Natl Acad Sci U S A 115, E302–E309. 10.1073/pnas.1716032115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Anderson MA, Ao Y, and Sofroniew MV (2014). Heterogeneity of reactive astrocytes. Neurosci Lett 565, 23–29. 10.1016/j.neulet.2013.12.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Patani R, Hardingham GE, and Liddelow SA (2023). Functional roles of reactive astrocytes in neuroinflammation and neurodegeneration. Nat Rev Neurol 19, 395–409. 10.1038/s41582-023-00822-1. [DOI] [PubMed] [Google Scholar]
- 132.Wheeler MA, Clark IC, Tjon EC, Li Z, Zandee SEJ, Couturier CP, Watson BR, Scalisi G, Alkwai S, Rothhammer V, et al. (2020). MAFG-driven astrocytes promote CNS inflammation. Nature 578, 593–599. 10.1038/s41586-020-1999-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Sun D, Peng H, and Wu Z (2022). Establishment and Analysis of a Combined Diagnostic Model of Alzheimer’s Disease With Random Forest and Artificial Neural Network. Front Aging Neurosci 14, 921906. 10.3389/fnagi.2022.921906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Wang P, Wang X, Qiao K, Zhang Y, Nie Q, Cui J, Sun J, and Li L (2023). Reduced SUMOylation of Nrf2 signaling contributes to its inhibition induced by amyloid-beta. Neurosci Lett 799, 137118. 10.1016/j.neulet.2023.137118. [DOI] [PubMed] [Google Scholar]
- 135.Wang X, Zhang Y, Wan X, Guo C, Cui J, Sun J, and Li L (2020). Responsive Expression of MafF to beta-Amyloid-Induced Oxidative Stress. Dis Markers 2020, 8861358. 10.1155/2020/8861358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Kataoka K, Igarashi K, Itoh K, Fujiwara KT, Noda M, Yamamoto M, and Nishizawa M (1995). Small Maf proteins heterodimerize with Fos and may act as competitive repressors of the NF-E2 transcription factor. Mol Cell Biol 15, 2180–2190. 10.1128/MCB.15.4.2180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Katsuoka F, and Yamamoto M (2016). Small Maf proteins (MafF, MafG, MafK): History, structure and function. Gene 586, 197–205. 10.1016/j.gene.2016.03.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Clark IC, Wheeler MA, Lee HG, Li Z, Sanmarco LM, Thaploo S, Polonio CM, Shin SW, Scalisi G, Henry AR, et al. (2023). Identification of astrocyte regulators by nucleic acid cytometry. Nature 614, 326–333. 10.1038/s41586-022-05613-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Sims SG, Cisney RN, Lipscomb MM, and Meares GP (2022). The role of endoplasmic reticulum stress in astrocytes. Glia 70, 5–19. 10.1002/glia.24082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Wheeler MA, Jaronen M, Covacu R, Zandee SEJ, Scalisi G, Rothhammer V, Tjon EC, Chao CC, Kenison JE, Blain M, et al. (2019). Environmental Control of Astrocyte Pathogenic Activities in CNS Inflammation. Cell 176, 581–596 e518. 10.1016/j.cell.2018.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Clayton BLL, Barbar L, Sapar M, Rusielewicz T, Kalpana K, Migliori B, Team NGSCA, Paull D, Brenner K, Moroziewicz D, et al. (2023). Patient iPSC models reveal glia-intrinsic phenotypes in multiple sclerosis. bioRxiv. 10.1101/2023.08.01.551553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Lee HG, Rone JM, Li Z, Akl CF, Shin SW, Lee JH, Flausino LE, Pernin F, Chao CC, Kleemann KL, et al. (2024). Disease-associated astrocyte epigenetic memory promotes CNS pathology. bioRxiv. 10.1101/2024.01.04.574196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Bjornevik K, Cortese M, Healy BC, Kuhle J, Mina MJ, Leng Y, Elledge SJ, Niebuhr DW, Scher AI, Munger KL, and Ascherio A (2022). Longitudinal analysis reveals high prevalence of Epstein-Barr virus associated with multiple sclerosis. Science 375, 296–301. 10.1126/science.abj8222. [DOI] [PubMed] [Google Scholar]
- 144.Robinson WH, and Steinman L (2022). Epstein-Barr virus and multiple sclerosis. Science 375, 264–265. 10.1126/science.abm7930. [DOI] [PubMed] [Google Scholar]
- 145.Sanmarco LM, Polonio CM, Wheeler MA, and Quintana FJ (2021). Functional immune cell-astrocyte interactions. J Exp Med 218. 10.1084/jem.20202715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Li S, Fang Y, Zhang Y, Song M, Zhang X, Ding X, Yao H, Chen M, Sun Y, Ding J, et al. (2022). Microglial NLRP3 inflammasome activates neurotoxic astrocytes in depression-like mice. Cell Rep 41, 111532. 10.1016/j.celrep.2022.111532. [DOI] [PubMed] [Google Scholar]
- 147.Guttikonda SR, Sikkema L, Tchieu J, Saurat N, Walsh RM, Harschnitz O, Ciceri G, Sneeboer M, Mazutis L, Setty M, et al. (2021). Fully defined human pluripotent stem cell-derived microglia and tri-culture system model C3 production in Alzheimer’s disease. Nat Neurosci 24, 343–354. 10.1038/s41593-020-00796-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Clark IC, Gutierrez-Vazquez C, Wheeler MA, Li Z, Rothhammer V, Linnerbauer M, Sanmarco LM, Guo L, Blain M, Zandee SEJ, et al. (2021). Barcoded viral tracing of single-cell interactions in central nervous system inflammation. Science 372. 10.1126/science.abf1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Stone S, Jamison S, Yue Y, Durose W, Schmidt-Ullrich R, and Lin W (2017). NF-kappaB Activation Protects Oligodendrocytes against Inflammation. J Neurosci 37, 9332–9344. 10.1523/JNEUROSCI.1608-17.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Lei Z, Yue Y, Stone S, Wu S, and Lin W (2020). NF-kappaB Activation Accounts for the Cytoprotective Effects of PERK Activation on Oligodendrocytes during EAE. J Neurosci 40, 6444–6456. 10.1523/JNEUROSCI.1156-20.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Chao CC, Gutierrez-Vazquez C, Rothhammer V, Mayo L, Wheeler MA, Tjon EC, Zandee SEJ, Blain M, de Lima KA, Takenaka MC, et al. (2019). Metabolic Control of Astrocyte Pathogenic Activity via cPLA2-MAVS. Cell 179, 1483–1498 e1422. 10.1016/j.cell.2019.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Ponath G, Lincoln MR, Levine-Ritterman M, Park C, Dahlawi S, Mubarak M, Sumida T, Airas L, Zhang S, Isitan C, et al. (2018). Enhanced astrocyte responses are driven by a genetic risk allele associated with multiple sclerosis. Nat Commun 9, 5337. 10.1038/s41467-018-07785-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Wheeler MA, Clark IC, Lee HG, Li Z, Linnerbauer M, Rone JM, Blain M, Akl CF, Piester G, Giovannoni F, et al. (2023). Droplet-based forward genetic screening of astrocyte-microglia cross-talk. Science 379, 1023–1030. 10.1126/science.abq4822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Housley WJ, Fernandez SD, Vera K, Murikinati SR, Grutzendler J, Cuerdon N, Glick L, De Jager PL, Mitrovic M, Cotsapas C, and Hafler DA (2015). Genetic variants associated with autoimmunity drive NFkappaB signaling and responses to inflammatory stimuli. Sci Transl Med 7, 291ra293. 10.1126/scitranslmed.aaa9223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.International Multiple Sclerosis Genetics, C. (2019). Multiple sclerosis genomic map implicates peripheral immune cells and microglia in susceptibility. Science 365. 10.1126/science.aav7188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Mead RJ, Singhrao SK, Neal JW, Lassmann H, and Morgan BP (2002). The membrane attack complex of complement causes severe demyelination associated with acute axonal injury. J Immunol 168, 458–465. 10.4049/jimmunol.168.1.458. [DOI] [PubMed] [Google Scholar]
- 157.Tassoni A, Farkhondeh V, Itoh Y, Itoh N, Sofroniew MV, and Voskuhl RR (2019). The astrocyte transcriptome in EAE optic neuritis shows complement activation and reveals a sex difference in astrocytic C3 expression. Sci Rep 9, 10010. 10.1038/s41598-019-46232-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Jin J, Smith MD, Kersbergen CJ, Kam TI, Viswanathan M, Martin K, Dawson TM, Dawson VL, Zack DJ, Whartenby K, and Calabresi PA (2019). Glial pathology and retinal neurotoxicity in the anterior visual pathway in experimental autoimmune encephalomyelitis. Acta Neuropathol Commun 7, 125. 10.1186/s40478-019-0767-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Xu L, Xu H, Chen S, Jiang W, Afridi SK, Wang Y, Ren X, Zhao Y, Lai S, Qiu X, et al. (2023). Inhibition of complement C3 signaling ameliorates locomotor and visual dysfunction in autoimmune inflammatory diseases. Mol Ther 31, 2715–2733. 10.1016/j.ymthe.2023.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Michailidou I, Willems JG, Kooi EJ, van Eden C, Gold SM, Geurts JJ, Baas F, Huitinga I, and Ramaglia V (2015). Complement C1q-C3-associated synaptic changes in multiple sclerosis hippocampus. Ann Neurol 77, 1007–1026. 10.1002/ana.24398. [DOI] [PubMed] [Google Scholar]
- 161.Dejanovic B, Wu T, Tsai MC, Graykowski D, Gandham VD, Rose CM, Bakalarski CE, Ngu H, Wang Y, Pandey S, et al. (2022). Complement C1q-dependent excitatory and inhibitory synapse elimination by astrocytes and microglia in Alzheimer’s disease mouse models. Nat Aging 2, 837–850. 10.1038/s43587-022-00281-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Werneburg S, Jung J, Kunjamma RB, Ha SK, Luciano NJ, Willis CM, Gao G, Biscola NP, Havton LA, Crocker SJ, et al. (2020). Targeted Complement Inhibition at Synapses Prevents Microglial Synaptic Engulfment and Synapse Loss in Demyelinating Disease. Immunity 52, 167–182 e167. 10.1016/j.immuni.2019.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Rajendran L, and Paolicelli RC (2018). Microglia-Mediated Synapse Loss in Alzheimer’s Disease. J Neurosci 38, 2911–2919. 10.1523/JNEUROSCI.1136-17.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Hammond JW, Bellizzi MJ, Ware C, Qiu WQ, Saminathan P, Li H, Luo S, Ma SA, Li Y, and Gelbard HA (2020). Complement-dependent synapse loss and microgliosis in a mouse model of multiple sclerosis. Brain Behav Immun 87, 739–750. 10.1016/j.bbi.2020.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Wu C, Yang L, Youngblood H, Liu TC, and Duan R (2022). Microglial SIRPalpha Deletion Facilitates Synapse Loss in Preclinical Models of Neurodegeneration. Neurosci Bull 38, 232–234. 10.1007/s12264-021-00795-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Eyo U, and Molofsky AV (2023). Defining microglial-synapse interactions. Science 381, 1155–1156. 10.1126/science.adh7906. [DOI] [PubMed] [Google Scholar]
- 167.Lindhout IA, Murray TE, Richards CM, and Klegeris A (2021). Potential neurotoxic activity of diverse molecules released by microglia. Neurochem Int 148, 105117. 10.1016/j.neuint.2021.105117. [DOI] [PubMed] [Google Scholar]
- 168.Murray TE, Richards CM, Robert-Gostlin VN, Bernath AK, Lindhout IA, and Klegeris A (2022). Potential neurotoxic activity of diverse molecules released by astrocytes. Brain Res Bull 189, 80–101. 10.1016/j.brainresbull.2022.08.015. [DOI] [PubMed] [Google Scholar]
- 169.Probert L, Akassoglou K, Kassiotis G, Pasparakis M, Alexopoulou L, and Kollias G (1997). TNF-alpha transgenic and knockout models of CNS inflammation and degeneration. J Neuroimmunol 72, 137–141. 10.1016/s0165-5728(96)00184-1. [DOI] [PubMed] [Google Scholar]
- 170.Pegoretti V, Baron W, Laman JD, and Eisel ULM (2018). Selective Modulation of TNF-TNFRs Signaling: Insights for Multiple Sclerosis Treatment. Front Immunol 9, 925. 10.3389/fimmu.2018.00925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Akassoglou K, Probert L, Kontogeorgos G, and Kollias G (1997). Astrocyte-specific but not neuron-specific transmembrane TNF triggers inflammation and degeneration in the central nervous system of transgenic mice. J Immunol 158, 438–445. [PubMed] [Google Scholar]
- 172.McCoy MK, Ruhn KA, Blesch A, and Tansey MG (2011). TNF: a key neuroinflammatory mediator of neurotoxicity and neurodegeneration in models of Parkinson’s disease. Adv Exp Med Biol 691, 539–540. 10.1007/978-1-4419-6612-4_56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Guttenplan KA, Weigel MK, Prakash P, Wijewardhane PR, Hasel P, Rufen-Blanchette U, Munch AE, Blum JA, Fine J, Neal MC, et al. (2021). Neurotoxic reactive astrocytes induce cell death via saturated lipids. Nature 599, 102–107. 10.1038/s41586-021-03960-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Dong Y, D’Mello C, Pinsky W, Lozinski BM, Kaushik DK, Ghorbani S, Moezzi D, Brown D, Melo FC, Zandee S, et al. (2021). Oxidized phosphatidylcholines found in multiple sclerosis lesions mediate neurodegeneration and are neutralized by microglia. Nat Neurosci 24, 489–503. 10.1038/s41593-021-00801-z. [DOI] [PubMed] [Google Scholar]
- 175.Xue S, Lozinski BM, Ghorbani S, Ta K, D’Mello C, Yong VW, and Dong Y (2023). Elevated Galectin-3 Is Associated with Aging, Multiple Sclerosis, and Oxidized Phosphatidylcholine-Induced Neurodegeneration. J Neurosci 43, 4725–4737. 10.1523/JNEUROSCI.2312-22.2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Krauss R, Bosanac T, Devraj R, Engber T, and Hughes RO (2020). Axons Matter: The Promise of Treating Neurodegenerative Disorders by Targeting SARM1-Mediated Axonal Degeneration. Trends Pharmacol Sci 41, 281–293. 10.1016/j.tips.2020.01.006. [DOI] [PubMed] [Google Scholar]
- 177.Lai MY, Li J, Zhang XX, Wu W, Li ZP, Sun ZX, Zhao MY, Yang DM, Wang DD, Li W, et al. (2022). SARM1 participates in axonal degeneration and mitochondrial dysfunction in prion disease. Neural Regen Res 17, 2293–2299. 10.4103/1673-5374.337051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Liu H, Zhang J, Xu X, Lu S, Yang D, Xie C, Jia M, Zhang W, Jin L, Wang X, et al. (2021). SARM1 promotes neuroinflammation and inhibits neural regeneration after spinal cord injury through NF-kappaB signaling. Theranostics 11, 4187–4206. 10.7150/thno.49054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Jin L, Zhang J, Hua X, Xu X, Li J, Wang J, Wang M, Liu H, Qiu H, Chen M, et al. (2022). Astrocytic SARM1 promotes neuroinflammation and axonal demyelination in experimental autoimmune encephalomyelitis through inhibiting GDNF signaling. Cell Death Dis 13, 759. 10.1038/s41419-022-05202-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Zhang J, Jin L, Hua X, Wang M, Wang J, Xu X, Liu H, Qiu H, Sun H, Dong T, et al. (2023). SARM1 promotes the neuroinflammation and demyelination through IGFBP2/NFkappaB pathway in experimental autoimmune encephalomyelitis mice. Acta Physiol (Oxf) 238, e13974. 10.1111/apha.13974. [DOI] [PubMed] [Google Scholar]
- 181.Viar K, Njoku D, Secor McVoy J, and Oh U (2020). Sarm1 knockout protects against early but not late axonal degeneration in experimental allergic encephalomyelitis. PLoS One 15, e0235110. 10.1371/journal.pone.0235110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Arredondo C, Cefaliello C, Dyrda A, Jury N, Martinez P, Diaz I, Amaro A, Tran H, Morales D, Pertusa M, et al. (2022). Excessive release of inorganic polyphosphate by ALS/FTD astrocytes causes non-cell-autonomous toxicity to motoneurons. Neuron 110, 1656–1670 e1612. 10.1016/j.neuron.2022.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Lassmann H (2018). Multiple Sclerosis Pathology. Cold Spring Harb Perspect Med 8. 10.1101/cshperspect.a028936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Fitzgerald KC, Smith MD, Kim S, Sotirchos ES, Kornberg MD, Douglas M, Nourbakhsh B, Graves J, Rattan R, Poisson L, et al. (2021). Multi-omic evaluation of metabolic alterations in multiple sclerosis identifies shifts in aromatic amino acid metabolism. Cell Rep Med 2, 100424. 10.1016/j.xcrm.2021.100424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Bhargava P, Fitzgerald KC, Calabresi PA, and Mowry EM (2017). Metabolic alterations in multiple sclerosis and the impact of vitamin D supplementation. JCI Insight 2. 10.1172/jci.insight.95302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Sangineto M, Ciarnelli M, Cassano T, Radesco A, Moola A, Bukke VN, Romano A, Villani R, Kanwal H, Capitanio N, et al. (2023). Metabolic reprogramming in inflammatory microglia indicates a potential way of targeting inflammation in Alzheimer’s disease. Redox Biol 66, 102846. 10.1016/j.redox.2023.102846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Pamies D, Sartori C, Schvartz D, Gonzalez-Ruiz V, Pellerin L, Nunes C, Tavel D, Maillard V, Boccard J, Rudaz S, et al. (2021). Neuroinflammatory Response to TNFalpha and IL1beta Cytokines Is Accompanied by an Increase in Glycolysis in Human Astrocytes In Vitro. Int J Mol Sci 22. 10.3390/ijms22084065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Robb JL, Hammad NA, Weightman Potter PG, Chilton JK, Beall C, and Ellacott KLJ (2020). The metabolic response to inflammation in astrocytes is regulated by nuclear factor-kappa B signaling. Glia 68, 2246–2263. 10.1002/glia.23835. [DOI] [PubMed] [Google Scholar]
- 189.Way SW, and Popko B (2016). Harnessing the integrated stress response for the treatment of multiple sclerosis. Lancet Neurol 15, 434–443. 10.1016/S1474-4422(15)00381-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Chen Y, Podojil JR, Kunjamma RB, Jones J, Weiner M, Lin W, Miller SD, and Popko B (2019). Sephin1, which prolongs the integrated stress response, is a promising therapeutic for multiple sclerosis. Brain 142, 344–361. 10.1093/brain/awy322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Way SW, Podojil JR, Clayton BL, Zaremba A, Collins TL, Kunjamma RB, Robinson AP, Brugarolas P, Miller RH, Miller SD, and Popko B (2015). Pharmaceutical integrated stress response enhancement protects oligodendrocytes and provides a potential multiple sclerosis therapeutic. Nat Commun 6, 6532. 10.1038/ncomms7532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Filippi M, Amato MP, Centonze D, Gallo P, Gasperini C, Inglese M, Patti F, Pozzilli C, Preziosa P, and Trojano M (2022). Early use of high-efficacy disease-modifying therapies makes the difference in people with multiple sclerosis: an expert opinion. J Neurol 269, 5382–5394. 10.1007/s00415-022-11193-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Barres BA, Schmid R, Sendnter M, and Raff MC (1993). Multiple extracellular signals are required for long-term oligodendrocyte survival. Development 118, 283–295. 10.1242/dev.118.1.283. [DOI] [PubMed] [Google Scholar]
- 194.Gharagozloo M, Bannon R, and Calabresi PA (2022). Breaking the barriers to remyelination in multiple sclerosis. Curr Opin Pharmacol 63, 102194. 10.1016/j.coph.2022.102194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Mi S, Lee X, Shao Z, Thill G, Ji B, Relton J, Levesque M, Allaire N, Perrin S, Sands B, et al. (2004). LINGO-1 is a component of the Nogo-66 receptor/p75 signaling complex. Nat Neurosci 7, 221–228. 10.1038/nn1188. [DOI] [PubMed] [Google Scholar]
- 196.Mi S, Hu B, Hahm K, Luo Y, Kam Hui ES, Yuan Q, Wong WM, Wang L, Su H, Chu TH, et al. (2007). LINGO-1 antagonist promotes spinal cord remyelination and axonal integrity in MOG-induced experimental autoimmune encephalomyelitis. Nat Med 13, 1228–1233. 10.1038/nm1664. [DOI] [PubMed] [Google Scholar]
- 197.Rudick RA, Mi S, and Sandrock AW Jr. (2008). LINGO-1 antagonists as therapy for multiple sclerosis: in vitro and in vivo evidence. Expert Opin Biol Ther 8, 1561–1570. 10.1517/14712598.8.10.1561. [DOI] [PubMed] [Google Scholar]
- 198.Quan Y, Wu Y, Zhan Z, Yang Y, Chen X, Wu K, and Yu M (2020). Inhibition of the leucine-rich repeat protein lingo-1 enhances RGC survival in optic nerve injury. Exp Ther Med 19, 619–629. 10.3892/etm.2019.8250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Moradbeygi K, Parviz M, Rezaeizadeh H, Zargaran A, Sahraian MA, Mehrabadi S, Nikbakhtzadeh M, and Zahedi E (2021). Anti-LINGO-1 improved remyelination and neurobehavioral deficit in cuprizone-induced demyelination. Iran J Basic Med Sci 24, 900–907. 10.22038/ijbms.2021.53531.12043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Mi S, Miller RH, Tang W, Lee X, Hu B, Wu W, Zhang Y, Shields CB, Zhang Y, Miklasz S, et al. (2009). Promotion of central nervous system remyelination by induced differentiation of oligodendrocyte precursor cells. Ann Neurol 65, 304–315. 10.1002/ana.21581. [DOI] [PubMed] [Google Scholar]
- 201.Cadavid D, Balcer L, Galetta S, Aktas O, Ziemssen T, Vanopdenbosch L, Frederiksen J, Skeen M, Jaffe GJ, Butzkueven H, et al. (2017). Safety and efficacy of opicinumab in acute optic neuritis (RENEW): a randomised, placebo-controlled, phase 2 trial. Lancet Neurol 16, 189–199. 10.1016/S1474-4422(16)30377-5. [DOI] [PubMed] [Google Scholar]
- 202.Cadavid D, Mellion M, Hupperts R, Edwards KR, Calabresi PA, Drulovic J, Giovannoni G, Hartung HP, Arnold DL, Fisher E, et al. (2019). Safety and efficacy of opicinumab in patients with relapsing multiple sclerosis (SYNERGY): a randomised, placebo-controlled, phase 2 trial. Lancet Neurol 18, 845–856. 10.1016/S1474-4422(19)30137-1. [DOI] [PubMed] [Google Scholar]
- 203.Mei F, Fancy SPJ, Shen YA, Niu J, Zhao C, Presley B, Miao E, Lee S, Mayoral SR, Redmond SA, et al. (2014). Micropillar arrays as a high-throughput screening platform for therapeutics in multiple sclerosis. Nat Med 20, 954–960. 10.1038/nm.3618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Li Z, He Y, Fan S, and Sun B (2015). Clemastine rescues behavioral changes and enhances remyelination in the cuprizone mouse model of demyelination. Neurosci Bull 31, 617–625. 10.1007/s12264-015-1555-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Yamazaki R, Osanai Y, Kouki T, Huang JK, and Ohno N (2023). Pharmacological treatment promoting remyelination enhances motor function after internal capsule demyelination in mice. Neurochem Int 164, 105505. 10.1016/j.neuint.2023.105505. [DOI] [PubMed] [Google Scholar]
- 206.Green AJ, Gelfand JM, Cree BA, Bevan C, Boscardin WJ, Mei F, Inman J, Arnow S, Devereux M, Abounasr A, et al. (2017). Clemastine fumarate as a remyelinating therapy for multiple sclerosis (ReBUILD): a randomised, controlled, double-blind, crossover trial. Lancet 390, 2481–2489. 10.1016/S0140-6736(17)32346-2. [DOI] [PubMed] [Google Scholar]
- 207.Brown JWL, Cunniffe NG, Prados F, Kanber B, Jones JL, Needham E, Georgieva Z, Rog D, Pearson OR, Overell J, et al. (2021). Safety and efficacy of bexarotene in patients with relapsing-remitting multiple sclerosis (CCMR One): a randomised, double-blind, placebo-controlled, parallel-group, phase 2a study. Lancet Neurol 20, 709–720. 10.1016/S1474-4422(21)00179-4. [DOI] [PubMed] [Google Scholar]
- 208.Brown JWL, Prados F, Altmann DR, Kanber B, Stutters J, Cunniffe NG, Jones JL, Georgieva ZG, Needham EJ, Daruwalla C, et al. (2022). Remyelination varies between and within lesions in multiple sclerosis following bexarotene. Ann Clin Transl Neurol 9, 1626–1642. 10.1002/acn3.51662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Schwartzbach CJ, Grove RA, Brown R, Tompson D, Then Bergh F, and Arnold DL (2017). Lesion remyelinating activity of GSK239512 versus placebo in patients with relapsing-remitting multiple sclerosis: a randomised, single-blind, phase II study. J Neurol 264, 304–315. 10.1007/s00415-016-8341-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Chataway J, De Angelis F, Connick P, Parker RA, Plantone D, Doshi A, John N, Stutters J, MacManus D, Prados Carrasco F, et al. (2020). Efficacy of three neuroprotective drugs in secondary progressive multiple sclerosis (MS-SMART): a phase 2b, multiarm, double-blind, randomised placebo-controlled trial. Lancet Neurol 19, 214–225. 10.1016/S1474-4422(19)30485-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Huntemann N, Rolfes L, Pawlitzki M, Ruck T, Pfeuffer S, Wiendl H, and Meuth SG (2021). Failed, Interrupted, or Inconclusive Trials on Neuroprotective and Neuroregenerative Treatment Strategies in Multiple Sclerosis: Update 2015–2020. Drugs 81, 1031–1063. 10.1007/s40265-021-01526-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Kuhlmann T, Miron V, Cui Q, Wegner C, Antel J, and Bruck W (2008). Differentiation block of oligodendroglial progenitor cells as a cause for remyelination failure in chronic multiple sclerosis. Brain 131, 1749–1758. 10.1093/brain/awn096. [DOI] [PubMed] [Google Scholar]
- 213.Chang A, Tourtellotte WW, Rudick R, and Trapp BD (2002). Premyelinating oligodendrocytes in chronic lesions of multiple sclerosis. N Engl J Med 346, 165–173. 10.1056/NEJMoa010994. [DOI] [PubMed] [Google Scholar]
- 214.Wolswijk G (1998). Chronic stage multiple sclerosis lesions contain a relatively quiescent population of oligodendrocyte precursor cells. J Neurosci 18, 601–609. 10.1523/JNEUROSCI.18-02-00601.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Starost L, Lindner M, Herold M, Xu YKT, Drexler HCA, Hess K, Ehrlich M, Ottoboni L, Ruffini F, Stehling M, et al. (2020). Extrinsic immune cell-derived, but not intrinsic oligodendroglial factors contribute to oligodendroglial differentiation block in multiple sclerosis. Acta Neuropathol 140, 715–736. 10.1007/s00401-020-02217-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Petersen MA, Tognatta R, Meyer-Franke A, Bushong EA, Mendiola AS, Yan Z, Muthusamy A, Merlini M, Meza-Acevedo R, Cabriga B, et al. (2021). BMP receptor blockade overcomes extrinsic inhibition of remyelination and restores neurovascular homeostasis. Brain 144, 2291–2301. 10.1093/brain/awab106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Keough MB, Rogers JA, Zhang P, Jensen SK, Stephenson EL, Chen T, Hurlbert MG, Lau LW, Rawji KS, Plemel JR, et al. (2016). An inhibitor of chondroitin sulfate proteoglycan synthesis promotes central nervous system remyelination. Nat Commun 7, 11312. 10.1038/ncomms11312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Baxi EG, DeBruin J, Tosi DM, Grishkan IV, Smith MD, Kirby LA, Strasburger HJ, Fairchild AN, Calabresi PA, and Gocke AR (2015). Transfer of myelin-reactive th17 cells impairs endogenous remyelination in the central nervous system of cuprizone-fed mice. J Neurosci 35, 8626–8639. 10.1523/JNEUROSCI.3817-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Gharagozloo M, Galleguillos D, Jank L, Sotirchos ES, Smith MD, Garton T, Kumar S, Hussein O, Potluri S, Taylor M, et al. (2023). The Effects of NLY01, a Novel Glucagon-Like Peptide-1 Receptor Agonist, on Cuprizone-Induced Demyelination and Remyelination: Challenges and Future Perspectives. Neurotherapeutics 20, 1229–1240. 10.1007/s13311-023-01390-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Caprariello AV, Rogers JA, Morgan ML, Hoghooghi V, Plemel JR, Koebel A, Tsutsui S, Dunn JF, Kotra LP, Ousman SS, et al. (2018). Biochemically altered myelin triggers autoimmune demyelination. Proc Natl Acad Sci U S A 115, 5528–5533. 10.1073/pnas.1721115115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Zinger N, Ponath G, Sweeney E, Nguyen TD, Lo CH, Diaz I, Dimov A, Teng L, Zexter L, Comunale J, et al. (2022). Dimethyl Fumarate Reduces Inflammation in Chronic Active Multiple Sclerosis Lesions. Neurol Neuroimmunol Neuroinflamm 9. 10.1212/NXI.0000000000001138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Kramer J, Bar-Or A, Turner TJ, and Wiendl H (2023). Bruton tyrosine kinase inhibitors for multiple sclerosis. Nat Rev Neurol 19, 289–304. 10.1038/s41582-023-00800-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Hu X, Tomlinson S, and Barnum SR (2012). Targeted inhibition of complement using complement receptor 2-conjugated inhibitors attenuates EAE. Neurosci Lett 531, 35–39. 10.1016/j.neulet.2012.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Hu X, Holers VM, Thurman JM, Schoeb TR, Ramos TN, and Barnum SR (2013). Therapeutic inhibition of the alternative complement pathway attenuates chronic EAE. Mol Immunol 54, 302–308. 10.1016/j.molimm.2012.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Dubreuil P, Letard S, Ciufolini M, Gros L, Humbert M, Casteran N, Borge L, Hajem B, Lermet A, Sippl W, et al. (2009). Masitinib (AB1010), a potent and selective tyrosine kinase inhibitor targeting KIT. PLoS One 4, e7258. 10.1371/journal.pone.0007258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Vermersch P, Brieva-Ruiz L, Fox RJ, Paul F, Ramio-Torrenta L, Schwab M, Moussy A, Mansfield C, Hermine O, Maciejowski M, and Group ABS (2022). Efficacy and Safety of Masitinib in Progressive Forms of Multiple Sclerosis: A Randomized, Phase 3, Clinical Trial. Neurol Neuroimmunol Neuroinflamm 9. 10.1212/NXI.0000000000001148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Li H, Liu S, Han J, Li S, Gao X, Wang M, Zhu J, and Jin T (2021). Role of Toll-Like Receptors in Neuroimmune Diseases: Therapeutic Targets and Problems. Front Immunol 12, 777606. 10.3389/fimmu.2021.777606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Zheng C, Chen J, Chu F, Zhu J, and Jin T (2019). Inflammatory Role of TLR-MyD88 Signaling in Multiple Sclerosis. Front Mol Neurosci 12, 314. 10.3389/fnmol.2019.00314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Fox RJ, Coffey CS, Conwit R, Cudkowicz ME, Gleason T, Goodman A, Klawiter EC, Matsuda K, McGovern M, Naismith RT, et al. (2018). Phase 2 Trial of Ibudilast in Progressive Multiple Sclerosis. N Engl J Med 379, 846–855. 10.1056/NEJMoa1803583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Liu W, Shi LJ, and Li SG (2019). The Immunomodulatory Effect of Alpha-Lipoic Acid in Autoimmune Diseases. Biomed Res Int 2019, 8086257. 10.1155/2019/8086257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Spain R, Powers K, Murchison C, Heriza E, Winges K, Yadav V, Cameron M, Kim E, Horak F, Simon J, and Bourdette D (2017). Lipoic acid in secondary progressive MS: A randomized controlled pilot trial. Neurol Neuroimmunol Neuroinflamm 4, e374. 10.1212/NXI.0000000000000374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Chataway J, Schuerer N, Alsanousi A, Chan D, MacManus D, Hunter K, Anderson V, Bangham CR, Clegg S, Nielsen C, et al. (2014). Effect of high-dose simvastatin on brain atrophy and disability in secondary progressive multiple sclerosis (MS-STAT): a randomised, placebo-controlled, phase 2 trial. Lancet 383, 2213–2221. 10.1016/S0140-6736(13)62242-4. [DOI] [PubMed] [Google Scholar]
- 233.Bhargava P, Smith MD, Mische L, Harrington E, Fitzgerald KC, Martin K, Kim S, Reyes AA, Gonzalez-Cardona J, Volsko C, et al. (2020). Bile acid metabolism is altered in multiple sclerosis and supplementation ameliorates neuroinflammation. J Clin Invest 130, 3467–3482. 10.1172/JCI129401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Chen J, Chia N, Kalari KR, Yao JZ, Novotna M, Paz Soldan MM, Luckey DH, Marietta EV, Jeraldo PR, Chen X, et al. (2016). Multiple sclerosis patients have a distinct gut microbiota compared to healthy controls. Sci Rep 6, 28484. 10.1038/srep28484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Khalaf K, Tornese P, Cocco A, and Albanese A (2022). Tauroursodeoxycholic acid: a potential therapeutic tool in neurodegenerative diseases. Transl Neurodegener 11, 33. 10.1186/s40035-022-00307-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Zucchi E, Musazzi UM, Fedele G, Martinelli I, Gianferrari G, Simonini C, Fini N, Ghezzi A, Caputo M, Sette E, et al. (2023). Effect of tauroursodeoxycholic acid on survival and safety in amyotrophic lateral sclerosis: a retrospective population-based cohort study. EClinicalMedicine 65, 102256. 10.1016/j.eclinm.2023.102256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Ladakis DC, Harrison KL, Smith MD, Solem K, Gadani S, Jank L, Hwang S, Farhadi F, Dewey BE, Fitzgerald KC, et al. (2024). Bile acid metabolites predict multiple sclerosis progression and supplementation is safe in progressive disease. medRxiv. 10.1101/2024.01.17.24301393. [DOI] [Google Scholar]
- 238.Beyer BA, Fang M, Sadrian B, Montenegro-Burke JR, Plaisted WC, Kok BPC, Saez E, Kondo T, Siuzdak G, and Lairson LL (2018). Metabolomics-based discovery of a metabolite that enhances oligodendrocyte maturation. Nat Chem Biol 14, 22–28. 10.1038/nchembio.2517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Petersen MA, Ryu JK, Chang KJ, Etxeberria A, Bardehle S, Mendiola AS, Kamau-Devers W, Fancy SPJ, Thor A, Bushong EA, et al. (2017). Fibrinogen Activates BMP Signaling in Oligodendrocyte Progenitor Cells and Inhibits Remyelination after Vascular Damage. Neuron 96, 1003–1012 e1007. 10.1016/j.neuron.2017.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Stephenson EL, Zhang P, Ghorbani S, Wang A, Gu J, Keough MB, Rawji KS, Silva C, Yong VW, and Ling CC (2019). Targeting the Chondroitin Sulfate Proteoglycans: Evaluating Fluorinated Glucosamines and Xylosides in Screens Pertinent to Multiple Sclerosis. ACS Cent Sci 5, 1223–1234. 10.1021/acscentsci.9b00327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Yu Z, Jiang N, Su W, and Zhuo Y (2021). Necroptosis: A Novel Pathway in Neuroinflammation. Front Pharmacol 12, 701564. 10.3389/fphar.2021.701564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Jiang X, Stockwell BR, and Conrad M (2021). Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol 22, 266–282. 10.1038/s41580-020-00324-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Biswas D, Dawson VL, and Dawson TM (2022). Pharmacologic inhibition of MIF nuclease: A new treatment paradigm to treat cell death. Clin Transl Med 12, e1044. 10.1002/ctm2.1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Zelic M, Pontarelli F, Woodworth L, Zhu C, Mahan A, Ren Y, LaMorte M, Gruber R, Keane A, Loring P, et al. (2021). RIPK1 activation mediates neuroinflammation and disease progression in multiple sclerosis. Cell Rep 35, 109112. 10.1016/j.celrep.2021.109112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Van San E, Debruyne AC, Veeckmans G, Tyurina YY, Tyurin VA, Zheng H, Choi SM, Augustyns K, van Loo G, Michalke B, et al. (2023). Ferroptosis contributes to multiple sclerosis and its pharmacological targeting suppresses experimental disease progression. Cell Death Differ 30, 2092–2103. 10.1038/s41418-023-01195-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Farez MF, Quintana FJ, Gandhi R, Izquierdo G, Lucas M, and Weiner HL (2009). Toll-like receptor 2 and poly(ADP-ribose) polymerase 1 promote central nervous system neuroinflammation in progressive EAE. Nat Immunol 10, 958–964. 10.1038/ni.1775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Mitroshina EV, Saviuk M, and Vedunova MV (2022). Necroptosis in CNS diseases: Focus on astrocytes. Front Aging Neurosci 14, 1016053. 10.3389/fnagi.2022.1016053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Xu X, Sun B, and Zhao C (2023). Poly (ADP-Ribose) polymerase 1 and parthanatos in neurological diseases: From pathogenesis to therapeutic opportunities. Neurobiol Dis 187, 106314. 10.1016/j.nbd.2023.106314. [DOI] [PubMed] [Google Scholar]
- 249.Park H, Kam TI, Peng H, Chou SC, Mehrabani-Tabari AA, Song JJ, Yin X, Karuppagounder SS, Umanah GK, Rao AVS, et al. (2022). PAAN/MIF nuclease inhibition prevents neurodegeneration in Parkinson’s disease. Cell 185, 1943–1959 e1921. 10.1016/j.cell.2022.04.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Ryan SK, Zelic M, Han Y, Teeple E, Chen L, Sadeghi M, Shankara S, Guo L, Li C, Pontarelli F, et al. (2023). Microglia ferroptosis is regulated by SEC24B and contributes to neurodegeneration. Nat Neurosci 26, 12–26. 10.1038/s41593-022-01221-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Rayatpour A, Foolad F, Heibatollahi M, Khajeh K, and Javan M (2022). Ferroptosis inhibition by deferiprone, attenuates myelin damage and promotes neuroprotection in demyelinated optic nerve. Sci Rep 12, 19630. 10.1038/s41598-022-24152-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Martinez-Zamudio RI, and Ha HC (2014). PARP1 enhances inflammatory cytokine expression by alteration of promoter chromatin structure in microglia. Brain Behav 4, 552–565. 10.1002/brb3.239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Li Y, Xiao D, and Wang X (2022). The emerging roles of ferroptosis in cells of the central nervous system. Front Neurosci 16, 1032140. 10.3389/fnins.2022.1032140. [DOI] [PMC free article] [PubMed] [Google Scholar]