Group-I-mGluRs and Microglia in CNS: focus on ALS: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , ,

Microglia cells are the resident immune cells of the central nervous system. They act as the first-line immune guardians of nervous tissue and central drivers of neuroinflammation. Any homeostatic alteration that can compromise neuron and tissue integrity could activate microglia. 

  • microglia
  • reactive phenotype
  • group I metabotropic glutamate receptors
  • amyotrophic lateral sclerosis
  • mGluR5
  • disease associated microglia

1. Role of Microglia in the Central Nervous System

Microglia represent the immune cells resident in the central nervous system (CNS) and are, therefore, the primary immune defense of this central area of the human body. Several studies suggest that microglia mainly originate from the embryonic yolk sac, and this evidence was confirmed in rodent models and humans [1][2][3][4][5]. In mice, microglia precursors enter the CNS around day 9.5 of embryonic life (E9.5) through extravascular pathways, as the first cerebral capillaries appear only on day E10 and are soon closed off from the periphery by E13.5, due to the blood–brain barrier’s (BBB) formation. In humans, microglia penetrate the cerebral cortex by gestational week 4.5 (GW4.5); then, a second wave of microglia infiltration penetrates the embryonic brain via the vasculature at GW12–13 [6][7][8][9][10][11][12]. After entering the brain, microglial cells rapidly proliferate [11][13][14][15] and, by GW22, take on a ramified morphology, becoming fully mature by GW35. Early entry and subsequent brain colonization represent actual events in the initial brain development.
Microglia regulate the number of neural precursors, promote neuron survival, and are involved in the phagocytosis of damaged neurons, synaptic pruning, angiogenesis, synaptogenesis, and the maturation of neural circuits [6][11][16][17][18][19][20][21][22][23]. Microglia have a unique genetic signature with respect to the perivascular, meningeal, and choroidal macrophages in the CNS, probably due to macrophages’ presumed hematopoietic origin and developmental processes [24]. Similarly, pre-natal and post-natal microglia differ from adult microglia [25][26]. Microglia play critical physiological roles during CNS development. Resting microglial cells exhibit numerous ramifications that constantly monitor the surrounding microenvironment, thus maintaining CNS homeostasis by phagocytosing cellular debris [27][28]. Microglia play an essential role in synaptic remodeling through synaptic pruning to optimize neurotransmission processes and are directly involved in the formation and reorganization of neural networks and in providing trophic support to mature neurons [12][20][29].
Microglia can sense the CNS environment by monitoring the signaling pathway molecules that guarantee the physiological crosstalk between microglia and neurons, which is fundamental for the maintenance of cerebral homeostasis [12][30][31][32][33]. A minimal variation in the extracellular milieu composition allows microglia to respond by modulating neuronal activity. The mature homeostatic microglia phenotype progresses in a multiple-step process during CNS development and requires continuous instructions from the adult brain [34][35][36][37][38].
Furthermore, microglia are promptly activated in response to CNS stimuli or pathological conditions and undergo massive morphological and functional changes accompanied by rapid clonal proliferation [39][40][41], a process identified as microglia activation. Activated microglia change their morphology from branched to amoeboid, resembling macrophages circulating in the bloodstream, and migrate toward the lesion site [28][42]. Convergence at the injury site occurs in response to signal molecules released by damaged neurons [42]. Activated microglia-induced neurotoxic effects can occur due to the release of cytotoxic molecules, including pro-inflammatory mediators, such as tumor necrosis factor α (TNF-α) or interferon γ (INFγ), and free radicals, superoxide anion, or nitric oxide (NO), and constituents triggering oxidative stress [43]. Under specific conditions, activated microglia acquire a defined anti-inflammatory phenotype, thus releasing neuroprotective factors [44].
Microglia express membrane receptors for several neurotransmitters [45], allowing them to respond to various external stimuli that determine the cell status. Indeed, some neurotransmitters can influence the activation state of microglia, producing changes in membrane potential as well as in the intracellular calcium concentration, causing the release of cytokines and generating cell motility [45][46]. In homeostatic or resting conditions, microglia exhibit the so-called “surveillant” phenotype, characterized by small cell bodies, limited cellular mobility, and extensive and highly mobile branches to control the surrounding environment [28][42]. This quiescent state is commonly identified as neutral, or “M0” [28][47], and characterized by the low expression of surface markers typical of circulating macrophages, i.e., the common lymphocyte antigen (CD45) and major histocompatibility complex class II (MHCII).
As discussed above, microglia are extremely sensitive to changes in the environment, thus being rapidly activated after exposure to specific signals, such as growth factors, neurotransmitters, or cytokines, that indicate the presence of infection, trauma, neuronal damage, or inflammation [48][49]. Moreover, microglia classify pathogens by recognizing damage-associated molecular patterns (DAMPs), receptor patterns of exogenous microorganisms, or endogenous cells involved in the immune response. As for peripheral macrophages, according to the first proposed nomenclature, the microglia activation state includes at least two distinct phenotypes: M1, described as a pro-inflammatory and neurotoxic phenotype, and M2, also known as the “alternative activation phenotype” [50][51][52], with anti-inflammatory and neuroprotective properties. These two phenotypes differently respond to distinct signals from the microenvironment and, in turn, are involved in producing many effector molecules [53], promoting the transcription of genes that activate cellular defense mechanisms, including the release of inflammatory cytokines and chemokines [50][54].

However, this unequivocal classification is inconsistent with the vast repertoire of microglial phenotypes and core functions in different situations, i.e., development, plasticity, ageing, and diseases. As recently reported, considering the coexistence of multiple states, microglia phenotypes occurring in a specific condition should be characterized by more potent analysis tools than those presently applied, such as proteomic, metabolomic, transcriptomic, morphological, and epigenetic ones [55]. Accordingly, a new nomenclature is needed to define the microglia phenotype in each specific physio-pathological environment. This scenario is even more complex since microglia cells can acquire specific activation cellular patterns depending on the pathological environmental conditions in which microglia participate, thus leading to different and peculiar “disease-associated microglia” (DAM) phenotypes [56].

2. Glutamate Receptors Expressed in Microglia

The specific activities of microglia likely result from the cell state, regulated by different environmental stimuli, which in turn activate cellular structures, mainly receptors, that act as sensors of external messengers and trigger several intracellular signals with distinct biological functions [46]. The excellent recent literature reports the presence and the role of many microglia-expressed receptor families. Purinergic, serotoninergic, histaminergic, and cannabinoid receptors are the most relevant in tuning the microglia state by regulating their phenotypic characteristics and functions, including proliferation, branch motility, cytokine release, cell migration, and phagocytosis, in physiological [57][58][59][60][61] and pathological conditions, including neurodegenerative diseases [62][63][64][65][66][67][68]. Although supported by limited evidence in the literature, it is worth mentioning that microglia also express GABAergic, cholinergic, adrenergic, and dopaminergic receptors [45][69].
Glial cells, including microglia, widely express glutamatergic receptors, whose activation exerts numerous crucial effects on the glia themselves and glia–neuron interactions in physiological and pathological conditions [70][71]. Glutamate (GLU) is the primary excitatory amino acid neurotransmitter in the brain. Once released at the presynaptic level, it activates post-synaptic α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA), N-methyl D-aspartate (NMDA), and kainate ionotropic receptors to stimulate rapid synaptic transmission [72]. GLU also activates G-protein-coupled metabotropic receptors (mGluRs) with slower signaling transduction kinetics. mGluRs include eight subtypes classified into three groups, termed I, II, and III, based on the sequence homology, signal transduction mechanism, and pharmacological profile [73]. When activated by GLU, mGluRs can generate fine feedback mechanisms at the pre-synaptic level, inhibiting or potentiating the release of GLU itself or other neurotransmitters from heterologous nerve terminals [74][75][76][77]. At the same time, mGluRs regulate critical cellular mechanisms, such as post-synaptic excitatory or inhibitory currents or the activity of glial cells surrounding the synapse, namely astrocytes and microglia [78]. In addition to the numerous actions in glial cells, the activation of mGluRs mediates the interaction between glia and neurons [79][80]. The latter effects are complex and bidirectional, often depending on the implicated mGluR subtypes [80].
Microglia express both ionotropic and metabotropic GLU receptors. These receptors mediate the response to GLU and participate in neuroinflammation and neurodegeneration processes [69]. Microglial NMDA receptors trigger neuroinflammation and neuronal death [81] by also driving pro-inflammatory responses via poly(ADP-ribose) polymerase-1 (PARP-1)/transient receptor potential cation channel subfamily M member 2 (TRMP2) signaling [82]. Thus, the existence of microglial NMDA receptors further offers a link between inflammation and excitotoxicity. Moreover, AMPA and kainate receptor activation triggers microglia reactivity and motor neuron toxicity [83][84][85]. mGluRs in microglia are involved in neuroinflammation [86], acute and chronic neurological disorders, and neurodegenerative diseases [87].

3. Physio-Pathological Role of Group I Metabotropic GLU Receptors Expressed by Microglia

Metabotropic GLU receptors are organized into three groups, termed I, II, and III, overall including eight subtypes (mGlu1–8 receptors) [73]. Group I include mGlu1 and mGlu5 receptors (henceforth reported as mGluR1 or mGluR5), which couple to a Gq protein, resulting in the activation of phospholipase C (PLC), and inositol triphosphate (IP3) and diacylglycerol (DAG) production. Group I receptors are mainly located in the post-synaptic compartment, and their activation increases cellular excitability. Group II, including mGluR2 and mGluR3, and group III, including mGluR4 and mGluR6–8, couple to Gi/Go proteins and inhibit adenylate cyclase (AC) activity and cyclic adenosine monophosphate (AMP) formation. Due to their widespread expression throughout the nervous system and the modulation of the relevant mechanisms that they participate in, mGluRs represent promising therapeutic targets to shape the microglia phenotype [88][89]. Some mGluR ligands are currently under clinical development regarding the treatment of various disorders, such as X Fragile, schizophrenia, Parkinson’s disease (PD), L-dopa-induced dyskinesias, generalized anxiety disorder, and chronic pain [90][91][92].
Microglia cell lines and primary cultures from the cerebral cortex express mGluR5 mRNA and protein [87][93][94]. Although some evidence indicates that quiescent microglia in the healthy brain do not express mGluR5 [95], after spinal cord or head trauma, activated microglia in the vicinity of the lesion significantly express this receptor [87][96].
Evidence demonstrating the presence of mGluR1 and mGluR5 in microglia has been obtained using selective receptor agonists and antagonists. The expression of the mGluR5a mRNA and the stimulation of calcium signaling by the mGluR1/5 agonist trans-(1S,3R)-1-amino-1,3-cyclopentane dicarboxylic acid (1S,3R-ACPD) took place in cultured microglia, indicating the expression of the mGluR5a variant in these cells [93]. However, Whittemore et al. [97] obtained conflicting results since they found no intracellular calcium signaling modification in microglia stimulated with 1S,3R-ACPD. The reasons for this discrepancy need to be clarified.
Other group I mGluR agonists trigger the activation of PLC in microglia cultures, which leads, again, to the release of calcium and activation of protein kinase C (PKC) [98]. In turn, PKC activation can cause changes in rectifying potassium channel expression and they shape microglia from the ameboid to the ramified phenotype [99]. Signaling downstream group I mGluRs include mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase 1 (ERK1), and extracellular signal-regulated kinase 2 (ERK2), which are inhibited by selective mGluR5 and mGluR1 antagonists, such as 2-Methyl-6-(phenylethynyl)pyridine (MPEP) and 7-(Hydroxyimino)cyclopropan[b]chromen-1a-carboxylate ethyl ester (CPCCOEt [98][100]). In cultured microglia, the selective activation of mGluR5 by (RS)-2-chloro-5-hidroxyphenylglycine (CHPG), or the combination of the mixed mGluR1/5 agonist [(S)-3,5-Dihydroxyphenylglycine] (DHPG) and the selective mGluR1 antagonist CPCCOEt, attenuated the lipopolysaccharide (LPS) or INFγ-induced activation [94][101], also reducing the accumulation of ROS, the production of TNF-α, and the levels of iNOS with consequent NO release. Moreover, PLC and PKC inhibitors and calcium chelators attenuated the anti-inflammatory events following mGluR5 activation, suggesting that the mGluR5 activation in microglia involves the Gq protein signal transduction pathway [102].
Loane et al. [94] showed that microglia express functional mGluR5, whose activation decreases the release of inflammatory molecules and the impact on neurotoxicity. The inhibition of NADPH oxidase mediated the protective effects of mGluR5 activation in microglia [103], a mechanism of microglia-mediated neurotoxicity common to numerous neurodegenerative diseases [104].
Another molecular pathway linked to the beneficial effects of mGluR5 in microglia is the brain-derived neurotrophic factor/tyrosine-protein kinase B (BDNF/TrKB) cascade [105]. Indeed, the activation of mGluR5 by CHPG protects from oxygen–glucose deprivation (OGD) and reperfusion-induced cytotoxicity, apoptosis, the accumulation of ROS, and the release of inflammatory cytokines in the microglial BV2 cell line [105]. mGluR5 activation also triggers the protein kinase B/glycogen synthase kinase 3β/cAMP-response element binding protein (Akt/GSK-3β/CREB) pathway, resulting in the inhibition of GSK-3β expression, increased phosphorylation of CREB, and reduced expression of inflammation-related genes in microglia cells [106].
Although initially only mRNA coding for mGluR5 was found in microglia cells, and cultured microglia apparently do not express mGluR1, in fact, the mGluR1 subtype appears to be present in this cell population [93], even if less expressed with respect to mGluR5 [102]. In addition to microglia, mGluR1 is also expressed by several cells within the CNS, including neurons, meningeal cells, astrocytes, and T and B cells [93][107], and mGluR1 agonists boost T cell proliferation and promote the activation of the MAPK signaling cascade, increasing inflammation [108]. Additionally, mGluR1 immunoreactivity has been reported in a subset of the microglia/macrophage cell lineage in human multiple sclerosis (MS) lesions [109]. Therefore, mGluR1 antagonists might also have multiple therapeutic applications.

3.1. Role of Group I Metabotropic GLU Receptors expressed by Microglia cells in amyotrophic lateral sclerosis

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive loss of upper and lower motor neurons (MNs). In the early phase, symptoms are muscle weakness, followed by a gradual loss of muscle control and contraction, atrophy, and paralysis, which lead to death by respiratory failure [110]. Around 90% of cases of ALS are sporadic or unrelated to a specific etiological or hereditary genetic cause. However, 10% of patients are familial, attributable to specific transmissible genetic mutations [111][112].

Although the initial ALS approach was mainly focused on neurons, growing and unequivocal evidence indicates that non-neuronal cells also play a key role in the pathogenesis of ALS, thus contributing to the definition of ALS as a non-cell-autonomous disease [113][114][115][116][117][118]. Glial cells, such as astrocytes and microglia, can participate in the local inflammatory response with peripheral lymphocytes and macrophages. They acquire a reactive phenotype, migrate to the lesion site, proliferate, and secrete pro-inflammatory and neurotoxic mediators [119][120][121][122][123][124][125][126][127]. Glial activation modifies the expression of a wide range of soluble molecules, such as cytokines and chemokines, DAMPs, reactive nitrogen species (RNS), and ROS, giving rise to profound changes in fundamental aspects of the interactions between glia and neurons [128].

Microglia become reactive before the symptomatic phase of the disease [52][129], concomitantly with the first loss of neuromuscular junctions [130] and MNs [131].

Several findings indicate that, during ALS progression, microglia cells do not undergo a stage-dependent transition [132]; instead, they show the coexistence of the different phenotypes, thus producing a peculiar functional profile as a complex outcome of multiple regulation factors.

Differing from other pathological or traumatic conditions, the involvement of group I mGluRs in the modulation of the microglia phenotype in ALS is poorly documented. Nevertheless, mGluR1, mGluR5, and microglia cells are indeed a promising target for ALS treatment and other neurodegenerative diseases in which neuroinflammation plays a pivotal role [70][87][89][133]. In ALS patients, mGluR1 and mGluR5 mRNAs are abundantly expressed in the dorsal horn rather than in the ventral horn of the spinal cord. Of note, spared MNs express abundant mGluR5, while vulnerable MNs do not [134]. Aronica and Collaborators showed that mGluR1 and mGluR5 were highly represented in neuronal cells throughout the human spinal cord, with mGluR1 having high expression in ventral horn neurons. In contrast, intense mGluR5 immunoreactivity was observed in the dorsal horns [135]. This different CNS area- and cell-specific expression of group I mGluRs highlights an intriguing clue possibly linked to the selective vulnerability of MNs in ALS [136][137][138][139]. Regarding glial cells, only sparse astrocytes showed weak to moderate staining for mGluR1 and mGluR5 in the spinal cords of healthy patients [135][140]. In ALS patients, the mGluR1 and mGluR5 immunolabeling intensity increased in cells with an astroglia morphology in the grey and white matter. At the same time, their expression in neurons was comparable to that observed in healthy subjects [135][140].

Although the role of mGluR5 in regulating astrocyte function and their neurotoxic phenotype during ALS progression was largely investigated [141][142][143][144][145][146], there is only one paper describing mGluRs affecting microglia functionality [147]. Berger et al. examined in vitro the modulation of mGluRs expressed by microglial cells in two distinct models of inflammatory conditions, including microglia cell cultures obtained from rats expressing the SOD1G93A ALS-linked mutation. As expected, SOD1G93A microglia were characterized by increased neuroinflammation and enhanced reactivity. The results showed that the mGluR5 mRNA was upregulated in microglia cell cultures prepared from the brains of neonatal SOD1G93A rats. Interestingly, the exposure to LPS, mimicking an inflammatory environment, increased mGluR3 and decreased mGluR5 gene expression in both SOD1G93A and wtSOD1 microglia [147]. This evidence indicates that an inflammatory environment may trigger the opposite regulation of mGluR subtype gene expression. These events seem particularly robust in SOD1G93A microglia cultures. Thus, it will be crucial to consider the possible cell-specific receptor expression and localization when con-sidering these therapeutic targets in ALS.

Using the SOD1G93A mouse model, it has been first demonstrated that the expression and function of mGluR1 and mGluR5 were enhanced at glutamatergic synapses in the spinal cord at the early pre-symptomatic and late symptomatic stages of the disease [75][76]. These alterations could further exacerbate the excessive glutamatergic neurotransmission previously demonstrated in the spinal cords of SOD1G93A mice [148][149][150]. In subsequent studies, genetically halving mGluR1 and mGluR5, or ablating mGluR5, significantly ameliorated disease progression and survival probability in SODG93A mice [151][152][153], and produced a reduction in astrogliosis and microgliosis, always accompanied by positive outcomes for the ALS phenotype. In vivo, the beneficial effects can hardly be ascribed to a specific cell subtype; however, researchers postulated that the observed modulation of reactive astrocytes and microglia could represent a potential contribution to the improved MN survival [151][152][153]. Then, the effect of the chronic oral administration of the mGluR5 NAM CTEP [154]. Differing from the mGluR5 genetic ablation, the pharmacological treatment was started after symptom onset and maintained until the late symptomatic stage of the disease [154]. CTEP dose-dependently ameliorated the survival and clinical course in SOD1G93A mice. Of relevance, paralleling the genetic studies, chronic treatment with CTEP also reduced astrogliosis and microglia proliferation in the spinal cords of SOD1G93A mice, possibly contributing to the amelioration of the extracellular noxious milieu toward MNs, thus in turn reducing the disease severity.

Considering the dual role of microglia during ALS progression and the fact that blocking mGluR5 before or after disease onset, by genetic or pharmacological strategies, respectively, always ameliorated disease progression and reduced glial reactivity, uncertainty about the mixed effects of dampening mGluR5 in ALS arises. The negative modulation of group I mGluRs should differently affect astrocyte or microglia cells early in the pathology, when microglia probably possess an anti-inflammatory phenotype and astrocytes should start to be reactive, with respect to the late symptomatic stages, when both astrocytes and microglia are detrimental for MNs.

The studies mentioned above, including the microglia-specific effects of group I mGluR modulation, are worth considering in the potential therapeutic application of mGluR5-targeted drugs to be exploited for ALS and other neurodegenerative diseases characterized by glial activation and neuroinflammatory features. Due to the subtle modifications that the microglia phenotype may undergo during specific ALS stages, and the uncertain role played by mGluRs, the only way to shed light on this complex scenario would be to expand the studies by exploiting more powerful and “omic” approaches, including transcriptomics, proteomics, metabolomics, and epigenomics, besides those adopted till now.

The pharmacological or genetic modulation of group I mGluRs expressed by microglia cells represents an attractive multipotential therapeutic strategy for acute traumatic and chronic neurodegenerative disorders. Although the literature has frequently investigated the roles of mGluRs and related therapeutic approaches focusing on neurons, other cell types, including astrocytes and microglia, express these receptors. Many neuroprotective strategies aimed at modulating the aberrant reactive state of these cells have highlighted microglia as a new target to improve clinical outcomes in different pathological conditions, particularly neurodegenerative diseases characterized by prominent neuroinflammatory hallmarks. Group I mGluR modulation reduces inflammation, excitotoxicity, necroptosis, and apoptotic cell death.

Further collection of preclinical and clinical evidence will be essential to optimize group I mGluRs as multipotential targets to modulate the complex balance of microglia phenotypes in CNS disorders.

This entry is adapted from the peer-reviewed paper 10.3390/ijms24065240

References

  1. Alliot, F.; Godin, I.; Pessac, B. Microglia derive from progenitors, originating from the yolk sac, and which proliferate in the brain. Dev. Brain Res. 1999, 117, 145–152.
  2. Rezaie, P.; Patel, K.; Male, D.K. Microglia in the human fetal spinal cord--patterns of distribution, morphology and phenotype. Dev. Brain Res. 1999, 115, 71–81.
  3. Chan, W.Y.; Kohsaka, S.; Rezaie, P. The origin and cell lineage of microglia: New concepts. Brain Res. Rev. 2007, 53, 344–354.
  4. Schulz, C.; Gomez Perdiguero, E.; Chorro, L.; Szabo-Rogers, H.; Cagnard, N.; Kierdorf, K.; Prinz, M.; Wu, B.; Jacobsen, S.E.W.; Pollard, J.W.; et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 2012, 336, 86–90.
  5. Kierdorf, K.; Erny, D.; Goldmann, T.; Sander, V.; Schulz, C.; Perdiguero, E.G.; Wieghofer, P.; Heinrich, A.; Riemke, P.; Hölscher, C.; et al. Microglia emerge from erythromyeloid precursors via Pu.1- and Irf8-dependent pathways. Nat. Neurosci. 2013, 16, 273–280.
  6. Monier, A.; Adle-Biassette, H.; Delezoide, A.-L.; Evrard, P.; Gressens, P.; Verney, C. Entry and distribution of microglial cells in human embryonic and fetal cerebral cortex. J. Neuropathol. Exp. Neurol. 2007, 66, 372–382.
  7. Verney, C.; Monier, A.; Fallet-Bianco, C.; Gressens, P. Early microglial colonization of the human forebrain and possible involvement in periventricular white-matter injury of preterm infants. J. Anat. 2010, 217, 436–448.
  8. Kurz, H.; Christ, B. Embryonic CNS macrophages and microglia do not stem from circulating, but from extravascular precursors. Glia 1998, 22, 98–102.
  9. Navascués, J.; Calvente, R.; Marín-Teva, J.L.; Cuadros, M.A. Entry, dispersion and differentiation of microglia in the developing central nervous system. An. Acad. Bras. Cienc. 2000, 72, 91–102.
  10. Ginhoux, F.; Greter, M.; Leboeuf, M.; Nandi, S.; See, P.; Gokhan, S.; Mehler, M.F.; Conway, S.J.; Ng, L.G.; Stanley, E.R.; et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 2010, 330, 841–845.
  11. Swinnen, N.; Smolders, S.; Avila, A.; Notelaers, K.; Paesen, R.; Ameloot, M.; Brône, B.; Legendre, P.; Rigo, J.-M. Complex invasion pattern of the cerebral cortex bymicroglial cells during development of the mouse embryo. Glia 2013, 61, 150–163.
  12. Nayak, D.; Roth, T.L.; McGavern, D.B. Microglia development and function. Annu. Rev. Immunol. 2014, 32, 367–402.
  13. Kim, I.; Mlsna, L.M.; Yoon, S.; Le, B.; Yu, S.; Xu, D.; Koh, S. A postnatal peak in microglial development in the mouse hippocampus is correlated with heightened sensitivity to seizure triggers. Brain Behav. 2015, 5, e00403.
  14. Nikodemova, M.; Kimyon, R.S.; De, I.; Small, A.L.; Collier, L.S.; Watters, J.J. Microglial numbers attain adult levels after undergoing a rapid decrease in cell number in the third postnatal week. J. Neuroimmunol. 2015, 278, 280–288.
  15. Eyo, U.B.; Miner, S.A.; Weiner, J.A.; Dailey, M.E. Developmental changes in microglial mobilization are independent of apoptosis in the neonatal mouse hippocampus. Brain. Behav. Immun. 2016, 55, 49–59.
  16. Wakselman, S.; Béchade, C.; Roumier, A.; Bernard, D.; Triller, A.; Bessis, A. Developmental neuronal death in hippocampus requires the microglial CD11b integrin and DAP12 immunoreceptor. J. Neurosci. 2008, 28, 8138–8143.
  17. Schafer, D.P.; Lehrman, E.K.; Kautzman, A.G.; Koyama, R.; Mardinly, A.R.; Yamasaki, R.; Ransohoff, R.M.; Greenberg, M.E.; Barres, B.A.; Stevens, B. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 2012, 74, 691–705.
  18. Cunningham, C.L.; Martínez-Cerdeño, V.; Noctor, S.C. Microglia regulate the number of neural precursor cells in the developing cerebral cortex. J. Neurosci. 2013, 33, 4216–4233.
  19. Parkhurst, C.N.; Yang, G.; Ninan, I.; Savas, J.N.; Yates, J.R., 3rd; Lafaille, J.J.; Hempstead, B.L.; Littman, D.R.; Gan, W.-B. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 2013, 155, 1596–1609.
  20. Ueno, M.; Fujita, Y.; Tanaka, T.; Nakamura, Y.; Kikuta, J.; Ishii, M.; Yamashita, T. Layer V cortical neurons require microglial support for survival during postnatal development. Nat. Neurosci. 2013, 16, 543–551.
  21. Pont-Lezica, L.; Beumer, W.; Colasse, S.; Drexhage, H.; Versnel, M.; Bessis, A. Microglia shape corpus callosum axon tract fasciculation: Functional impact of prenatal inflammation. Eur. J. Neurosci. 2014, 39, 1551–1557.
  22. Squarzoni, P.; Oller, G.; Hoeffel, G.; Pont-Lezica, L.; Rostaing, P.; Low, D.; Bessis, A.; Ginhoux, F.; Garel, S. Microglia modulate wiring of the embryonic forebrain. Cell Rep. 2014, 8, 1271–1279.
  23. Hattori, Y. The behavior and functions of embryonic microglia. Anat. Sci. Int. 2022, 97, 1–14.
  24. Li, Q.; Barres, B.A. Microglia and macrophages in brain homeostasis and disease. Nat. Rev. Immunol. 2018, 18, 225–242.
  25. Matcovitch-Natan, O.; Winter, D.R.; Giladi, A.; Vargas Aguilar, S.; Spinrad, A.; Sarrazin, S.; Ben-Yehuda, H.; David, E.; Zelada González, F.; Perrin, P.; et al. Microglia development follows a stepwise program to regulate brain homeostasis. Science 2016, 353, aad8670.
  26. Yoo, H.-J.; Kwon, M.-S. Aged Microglia in Neurodegenerative Diseases: Microglia Lifespan and Culture Methods. Front. Aging Neurosci. 2021, 13, 766267.
  27. Neumann, H.; Kotter, M.R.; Franklin, R.J.M. Debris clearance by microglia: An essential link between degeneration and regeneration. Brain 2009, 132, 288–295.
  28. Nimmerjahn, A.; Kirchhoff, F.; Helmchen, F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 2005, 308, 1314–1318.
  29. Bessis, A.; Béchade, C.; Bernard, D.; Roumier, A. Microglial control of neuronal death and synaptic properties. Glia 2007, 55, 233–238.
  30. Ransohoff, R.M.; Perry, V.H. Microglial physiology: Unique stimuli, specialized responses. Annu. Rev. Immunol. 2009, 27, 119–145.
  31. Li, Y.; Du, X.-F.; Liu, C.-S.; Wen, Z.-L.; Du, J.-L. Reciprocal regulation between resting microglial dynamics and neuronal activity in vivo. Dev. Cell 2012, 23, 1189–1202.
  32. Eyo, U.B.; Dailey, M.E. Microglia: Key elements in neural development, plasticity, and pathology. J. Neuroimmune Pharmacol. Off. J. Soc. NeuroImmune Pharmacol. 2013, 8, 494–509.
  33. Eyo, U.B.; Wu, L.-J. Bidirectional microglia-neuron communication in the healthy brain. Neural Plast. 2013, 2013, 456857.
  34. Baxter, P.S.; Dando, O.; Emelianova, K.; He, X.; McKay, S.; Hardingham, G.E.; Qiu, J. Microglial identity and inflammatory responses are controlled by the combined effects of neurons and astrocytes. Cell Rep. 2021, 34, 108882.
  35. Bennett, F.C.; Bennett, M.L.; Yaqoob, F.; Mulinyawe, S.B.; Grant, G.A.; Hayden Gephart, M.; Plowey, E.D.; Barres, B.A. A Combination of Ontogeny and CNS Environment Establishes Microglial Identity. Neuron 2018, 98, 1170–1183.e8.
  36. Elmore, M.R.P.; Najafi, A.R.; Koike, M.A.; Dagher, N.N.; Spangenberg, E.E.; Rice, R.A.; Kitazawa, M.; Matusow, B.; Nguyen, H.; West, B.L.; et al. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron 2014, 82, 380–397.
  37. Gosselin, D.; Link, V.M.; Romanoski, C.E.; Fonseca, G.J.; Eichenfield, D.Z.; Spann, N.J.; Stender, J.D.; Chun, H.B.; Garner, H.; Geissmann, F.; et al. Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell 2014, 159, 1327–1340.
  38. Gosselin, D.; Skola, D.; Coufal, N.G.; Holtman, I.R.; Schlachetzki, J.C.M.; Sajti, E.; Jaeger, B.N.; O’Connor, C.; Fitzpatrick, C.; Pasillas, M.P.; et al. An environment-dependent transcriptional network specifies human microglia identity. Science 2017, 356, aal3222.
  39. Füger, P.; Hefendehl, J.K.; Veeraraghavalu, K.; Wendeln, A.-C.; Schlosser, C.; Obermüller, U.; Wegenast-Braun, B.M.; Neher, J.J.; Martus, P.; Kohsaka, S.; et al. Microglia turnover with aging and in an Alzheimer’s model via long-term in vivo single-cell imaging. Nat. Neurosci. 2017, 20, 1371–1376.
  40. Jordão, M.J.C.; Sankowski, R.; Brendecke, S.M.; Sagar; Locatelli, G.; Tai, Y.-H.; Tay, T.L.; Schramm, E.; Armbruster, S.; Hagemeyer, N.; et al. Single-cell profiling identifies myeloid cell subsets with distinct fates during neuroinflammation. Science 2019, 363, aat7554.
  41. Tay, T.L.; Mai, D.; Dautzenberg, J.; Fernández-Klett, F.; Lin, G.; Sagar; Datta, M.; Drougard, A.; Stempfl, T.; Ardura-Fabregat, A.; et al. A new fate mapping system reveals context-dependent random or clonal expansion of microglia. Nat. Neurosci. 2017, 20, 793–803.
  42. Davalos, D.; Grutzendler, J.; Yang, G.; Kim, J.V.; Zuo, Y.; Jung, S.; Littman, D.R.; Dustin, M.L.; Gan, W.-B. ATP mediates rapid microglial response to local brain injury in vivo. Nat. Neurosci. 2005, 8, 752–758.
  43. Block, M.L.; Hong, J.-S. Microglia and inflammation-mediated neurodegeneration: Multiple triggers with a common mechanism. Prog. Neurobiol. 2005, 76, 77–98.
  44. Fujita, Y.; Yamashita, T. Neuroprotective function of microglia in the developing brain. Neuronal Signal. 2021, 5, NS20200024.
  45. Kettenmann, H.; Hanisch, U.K.; Noda, M.; Verkhratsky, A. Physiology of microglia. Physiol. Rev. 2011, 91, 461–553.
  46. Pocock, J.M.; Kettenmann, H. Neurotransmitter receptors on microglia. Trends Neurosci. 2007, 30, 527–535.
  47. Timmerman, R.; Burm, S.M.; Bajramovic, J.J. An Overview of in vitro Methods to Study Microglia. Front. Cell. Neurosci. 2018, 12, 242.
  48. Lynch, M.A. The multifaceted profile of activated microglia. Mol. Neurobiol. 2009, 40, 139–156.
  49. Mosser, C.-A.; Baptista, S.; Arnoux, I.; Audinat, E. Microglia in CNS development: Shaping the brain for the future. Prog. Neurobiol. 2017, 149–150, 1–20.
  50. Colton, C.A. Heterogeneity of microglial activation in the innate immune response in the brain. J. Neuroimmune Pharmacol. Off. J. Soc. NeuroImmune Pharmacol. 2009, 4, 399–418.
  51. Colton, C.A.; Wilcock, D.M. Assessing activation states in microglia. CNS Neurol. Disord. Drug Targets 2010, 9, 174–191.
  52. Tang, Y.; Le, W. Differential Roles of M1 and M2 Microglia in Neurodegenerative Diseases. Mol. Neurobiol. 2016, 53, 1181–1194.
  53. Du, L.; Zhang, Y.; Chen, Y.; Zhu, J.; Yang, Y.; Zhang, H.-L. Role of Microglia in Neurological Disorders and Their Potentials as a Therapeutic Target. Mol. Neurobiol. 2017, 54, 7567–7584.
  54. Kigerl, K.A.; de Rivero Vaccari, J.P.; Dietrich, W.D.; Popovich, P.G.; Keane, R.W. Pattern recognition receptors and central nervous system repair. Exp. Neurol. 2014, 258, 5–16.
  55. Paolicelli, R.C.; Sierra, A.; Stevens, B.; Tremblay, M.-E.; Aguzzi, A.; Ajami, B.; Amit, I.; Audinat, E.; Bechmann, I.; Bennett, M.; et al. Microglia states and nomenclature: A field at its crossroads. Neuron 2022, 110, 3458–3483.
  56. Guo, S.; Wang, H.; Yin, Y. Microglia Polarization From M1 to M2 in Neurodegenerative Diseases. Front. Aging Neurosci. 2022, 14, 815347.
  57. Castellano, B.; Bosch-Queralt, M.; Almolda, B.; Villacampa, N.; González, B. Purine Signaling and Microglial Wrapping. Adv. Exp. Med. Biol. 2016, 949, 147–165.
  58. Calovi, S.; Mut-Arbona, P.; Sperlágh, B. Microglia and the Purinergic Signaling System. Neuroscience 2019, 405, 137–147.
  59. Franco, R.; Lillo, A.; Rivas-Santisteban, R.; Reyes-Resina, I.; Navarro, G. Microglial Adenosine Receptors: From Preconditioning to Modulating the M1/M2 Balance in Activated Cells. Cells 2021, 10, 1124.
  60. Krabbe, G.; Matyash, V.; Pannasch, U.; Mamer, L.; Boddeke, H.W.G.M.; Kettenmann, H. Activation of serotonin receptors promotes microglial injury-induced motility but attenuates phagocytic activity. Brain. Behav. Immun. 2012, 26, 419–428.
  61. Iida, T.; Yanai, K.; Yoshikawa, T. Histamine and Microglia. Curr. Top. Behav. Neurosci. 2022, 59, 241–259.
  62. Volonté, C.; Apolloni, S.; Parisi, C.; Amadio, S. Purinergic contribution to amyotrophic lateral sclerosis. Neuropharmacology 2016, 104, 180–193.
  63. Woods, L.T.; Ajit, D.; Camden, J.M.; Erb, L.; Weisman, G.A. Purinergic receptors as potential therapeutic targets in Alzheimer’s disease. Neuropharmacology 2016, 104, 169–179.
  64. El Oussini, H.; Bayer, H.; Scekic-Zahirovic, J.; Vercruysse, P.; Sinniger, J.; Dirrig-Grosch, S.; Dieterlé, S.; Echaniz-Laguna, A.; Larmet, Y.; Müller, K.; et al. Serotonin 2B receptor slows disease progression and prevents degeneration of spinal cord mononuclear phagocytes in amyotrophic lateral sclerosis. Acta Neuropathol. 2016, 131, 465–480.
  65. Barata-Antunes, S.; Cristóvão, A.C.; Pires, J.; Rocha, S.M.; Bernardino, L. Dual role of histamine on microglia-induced neurodegeneration. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 764–769.
  66. Mecha, M.; Carrillo-Salinas, F.J.; Feliú, A.; Mestre, L.; Guaza, C. Microglia activation states and cannabinoid system: Therapeutic implications. Pharmacol. Ther. 2016, 166, 40–55.
  67. Komorowska-Müller, J.A.; Schmöle, A.-C. CB2 Receptor in Microglia: The Guardian of Self-Control. Int. J. Mol. Sci. 2020, 22, 19.
  68. Duffy, S.S.; Hayes, J.P.; Fiore, N.T.; Moalem-Taylor, G. The cannabinoid system and microglia in health and disease. Neuropharmacology 2021, 190, 108555.
  69. Liu, H.; Leak, R.K.; Hu, X. Neurotransmitter receptors on microglia. Stroke Vasc. Neurol. 2016, 1, 52–58.
  70. Volonté, C.; Amadio, S.; Fabbrizio, P.; Apolloni, S. Functional microglia neurotransmitters in amyotrophic lateral sclerosis. Semin. Cell Dev. Biol. 2019, 94, 121–128.
  71. Zhang, X.; Wang, D.; Zhang, B.; Zhu, J.; Zhou, Z.; Cui, L. Regulation of microglia by glutamate and its signal pathway in neurodegenerative diseases. Drug Discov. Today 2020, 25, 1074–1085.
  72. Dingledine, R.; Borges, K.; Bowie, D.; Traynelis, S.F. The glutamate receptor ion channels. Pharmacol. Rev. 1999, 51, 7–61.
  73. Willard, S.S.; Koochekpour, S. Glutamate, Glutamate Receptors, and Downstream Signaling Pathways. Int. J. Biol. Sci. 2013, 9, 948.
  74. Bragina, L.; Bonifacino, T.; Bassi, S.; Milanese, M.; Bonanno, G.; Conti, F. Differential expression of metabotropic glutamate and GABA receptors at neocortical glutamatergic and GABAergic axon terminals. Front. Cell. Neurosci. 2015, 9, 345.
  75. Giribaldi, F.; Milanese, M.; Bonifacino, T.; Anna Rossi, P.I.; Di Prisco, S.; Pittaluga, A.; Tacchetti, C.; Puliti, A.; Usai, C.; Bonanno, G. Group I metabotropic glutamate autoreceptors induce abnormal glutamate exocytosis in a mouse model of amyotrophic lateral sclerosis. Neuropharmacology 2013, 66, 253–263.
  76. Bonifacino, T.; Rebosio, C.; Provenzano, F.; Torazza, C.; Balbi, M.; Milanese, M.; Raiteri, L.; Usai, C.; Fedele, E.; Bonanno, G. Enhanced function and overexpression of metabotropic glutamate receptors 1 and 5 in the spinal Cord of the SOD1G93A mouse model of amyotrophic lateral sclerosis during disease progression. Int. J. Mol. Sci. 2019, 20, 4552.
  77. Olivero, G.; Vergassola, M.; Cisani, F.; Roggeri, A.; Pittaluga, A. Presynaptic Release-regulating Metabotropic Glutamate Receptors: An Update. Curr. Neuropharmacol. 2020, 18, 655–672.
  78. Conn, P.J.; Pin, J.P. Pharmacology and functions of metabotropic glutamate receptors. Annu. Rev. Pharmacol. Toxicol. 1997, 37, 205–237.
  79. D’Antoni, S.; Berretta, A.; Bonaccorso, C.M.; Bruno, V.; Aronica, E.; Nicoletti, F.; Catania, M.V. Metabotropic glutamate receptors in glial cells. Neurochem. Res. 2008, 33, 2436–2443.
  80. Spampinato, S.F.; Copani, A.; Nicoletti, F.; Sortino, M.A.; Caraci, F. Metabotropic Glutamate Receptors in Glial Cells: A New Potential Target for Neuroprotection? Front. Mol. Neurosci. 2018, 11, 414.
  81. Kaindl, A.M.; Degos, V.; Peineau, S.; Gouadon, E.; Chhor, V.; Loron, G.; Le Charpentier, T.; Josserand, J.; Ali, C.; Vivien, D.; et al. Activation of microglial N-methyl-D-aspartate receptors triggers inflammation and neuronal cell death in the developing and mature brain. Ann. Neurol. 2012, 72, 536–549.
  82. Raghunatha, P.; Vosoughi, A.; Kauppinen, T.M.; Jackson, M.F. Microglial NMDA receptors drive pro-inflammatory responses via PARP-1/TRMP2 signaling. Glia 2020, 68, 1421–1434.
  83. Zhao, W.; Xie, W.; Le, W.; Beers, D.R.; He, Y.; Henkel, J.S.; Simpson, E.P.; Yen, A.A.; Xiao, Q.; Appel, S.H. Activated microglia initiate motor neuron injury by a nitric oxide and glutamate-mediated mechanism. J. Neuropathol. Exp. Neurol. 2004, 63, 964–977.
  84. Ceprian, M.; Fulton, D. Glial Cell AMPA Receptors in Nervous System Health, Injury and Disease. Int. J. Mol. Sci. 2019, 20, 2450.
  85. Zhang, X.-M.; Zhu, J. Kainic Acid-induced neurotoxicity: Targeting glial responses and glia-derived cytokines. Curr. Neuropharmacol. 2011, 9, 388–398.
  86. Fazio, F.; Ulivieri, M.; Volpi, C.; Gargaro, M.; Fallarino, F. Targeting metabotropic glutamate receptors for the treatment of neuroinflammation. Curr. Opin. Pharmacol. 2018, 38, 16–23.
  87. Byrnes, K.R.; Loane, D.J.; Faden, A.I. Metabotropic glutamate receptors as targets for multipotential treatment of neurological disorders. Neurother. J. Am. Soc. Exp. Neurother. 2009, 6, 94–107.
  88. Conn, P.J. Physiological roles and therapeutic potential of metabotropic glutamate receptors. Ann. N. Y. Acad. Sci. 2003, 1003, 12–21.
  89. Battaglia, G.; Bruno, V. Metabotropic glutamate receptor involvement in the pathophysiology of amyotrophic lateral sclerosis: New potential drug targets for therapeutic applications. Curr. Opin. Pharmacol. 2018, 38, 65–71.
  90. Nicoletti, F.; Bockaert, J.; Collingridge, G.L.; Conn, P.J.; Ferraguti, F.; Schoepp, D.D.; Wroblewski, J.T.; Pin, J.P. Metabotropic glutamate receptors: From the workbench to the bedside. Neuropharmacology 2011, 60, 1017–1041.
  91. Berry-Kravis, E.M.; Lindemann, L.; Jønch, A.E.; Apostol, G.; Bear, M.F.; Carpenter, R.L.; Crawley, J.N.; Curie, A.; Des Portes, V.; Hossain, F.; et al. Drug development for neurodevelopmental disorders: Lessons learned from fragile X syndrome. Nat. Rev. Drug Discov. 2018, 17, 280–299.
  92. Su, L.-D.; Wang, N.; Han, J.; Shen, Y. Group 1 Metabotropic Glutamate Receptors in Neurological and Psychiatric Diseases: Mechanisms and Prospective. Neurosci. Rev. J. Bringing Neurobiol. Neurol. Psychiatry 2022, 28, 453–468.
  93. Biber, K.; Laurie, D.J.; Berthele, A.; Sommer, B.; Tölle, T.R.; Gebicke-Härter, P.J.; Van Calker, D.; Boddeke, H.W.G.M. Expression and signaling of group I metabotropic glutamate receptors in astrocytes and microglia. J. Neurochem. 1999, 72, 1671–1680.
  94. Loane, D.J.; Stoica, B.A.; Pajoohesh-Ganji, A.; Byrnes, K.R.; Faden, A.I. Activation of metabotropic glutamate receptor 5 modulates microglial reactivity and neurotoxicity by inhibiting NADPH oxidase. J. Biol. Chem. 2009, 284, 15629–15639.
  95. Mudo, G.; Trovato-Salinaro, A.; Caniglia, G.; Cheng, Q.; Condorelli, D.F. Cellular localization of mGluR3 and mGluR5 mRNAs in normal and injured rat brain. Brain Res. 2007, 1149, 1–13.
  96. Drouin-Ouellet, J.; Brownell, A.-L.L.; Saint-Pierre, M.; Fasano, C.; Emond, V.; Trudeau, L.-E.E.; Lévesque, D.; Cicchetti, F. Neuroinflammation is associated with changes in glial mGluR5 expression and the development of neonatal excitotoxic lesions. Glia 2011, 59, 188–199.
  97. Whittemore, E.R.; Korotzer, A.R.; Etebari, A.; Cotman, C.W. Carbachol increases intracellular free calcium in cultured rat microglia. Brain Res. 1993, 621, 59–64.
  98. Karim, F.; Wang, C.C.; Gereau, R.W. 4th Metabotropic glutamate receptor subtypes 1 and 5 are activators of extracellular signal-regulated kinase signaling required for inflammatory pain in mice. J. Neurosci. 2001, 21, 3771–3779.
  99. Eder, C. Ion channels in microglia (brain macrophages). Am. J. Physiol. 1998, 275, C327–C342.
  100. Warwick, H.K.; Nahorski, S.R.; Challiss, R.A.J. Group I metabotropic glutamate receptors, mGlu1a and mGlu5a, couple to cyclic AMP response element binding protein (CREB) through a common Ca2+- and protein kinase C-dependent pathway. J. Neurochem. 2005, 93, 232–245.
  101. Farso, M.C.; O’Shea, R.D.; Beart, P.M. Evidence group I mGluR drugs modulate the activation profile of lipopolysaccharide-exposed microglia in culture. Neurochem. Res. 2009, 34, 1721–1728.
  102. Byrnes, K.R.; Stoica, B.; Loane, D.J.; Riccio, A.; Davis, M.I.; Faden, A.I. Metabotropic glutamate receptor 5 activation inhibits microglial associated inflammation and neurotoxicity. Glia 2009, 57, 550–560.
  103. Loane, D.J.; Stoica, B.A.; Byrnes, K.R.; Jeong, W.; Faden, A.I. Activation of mGluR5 and inhibition of NADPH oxidase improves functional recovery after traumatic brain injury. J. Neurotrauma 2013, 30, 403–412.
  104. Lull, M.E.; Block, M.L. Microglial activation and chronic neurodegeneration. Neurother. J. Am. Soc. Exp. Neurother. 2010, 7, 354–365.
  105. Ye, X.; Yu, L.; Zuo, D.; Zhang, L.; Zu, J.; Hu, J.; Tang, J.; Bao, L.; Cui, C.; Zhang, R.; et al. Activated mGluR5 protects BV2 cells against OGD/R induced cytotoxicity by modulating BDNF-TrkB pathway. Neurosci. Lett. 2017, 654, 70–79.
  106. Bhat, S.A.; Henry, R.J.; Blanchard, A.C.; Stoica, B.A.; Loane, D.J.; Faden, A.I. Enhanced Akt/GSK-3β/CREB signaling mediates the anti-inflammatory actions of mGluR5 positive allosteric modulators in microglia and following traumatic brain injury in male mice. J. Neurochem. 2021, 156, 225–248.
  107. Hermans, E.; Challiss, R.A. Structural, signalling and regulatory properties of the group I metabotropic glutamate receptors: Prototypic family C G-protein-coupled receptors. Biochem. J. 2001, 359, 465–484.
  108. Pacheco, R.; Ciruela, F.; Casadó, V.; Mallol, J.; Gallart, T.; Lluis, C.; Franco, R. Group I metabotropic glutamate receptors mediate a dual role of glutamate in T cell activation. J. Biol. Chem. 2004, 279, 33352–33358.
  109. Geurts, J.J.G.; Wolswijk, G.; Bö, L.; van der Valk, P.; Polman, C.H.; Troost, D.; Aronica, E. Altered expression patterns of group I and II metabotropic glutamate receptors in multiple sclerosis. Brain 2003, 126, 1755–1766.
  110. Brown, R.H.; Al-Chalabi, A. Amyotrophic Lateral Sclerosis. N. Engl. J. Med. 2017, 377, 162–172.
  111. Renton, A.E.; Chiò, A.; Traynor, B.J. State of play in amyotrophic lateral sclerosis genetics. Nat. Publ. Gr. 2013, 17, 17–23.
  112. Rowland, L.P.; Shneider, N.A. Amyotrophic lateral sclerosis. N. Engl. J. Med. 2001, 344, 1688–1700.
  113. Van Harten, A.C.M.M.; Phatnani, H.; Przedborski, S. Non-cell-autonomous pathogenic mechanisms in amyotrophic lateral sclerosis. Trends Neurosci. 2021, 44, 658–668.
  114. Li, L.; Acioglu, C.; Heary, R.F.; Elkabes, S. Role of astroglial toll-like receptors (TLRs) in central nervous system infections, injury and neurodegenerative diseases. Brain. Behav. Immun. 2021, 91, 740–755.
  115. Kim, C.; Yousefian-Jazi, A.; Choi, S.-H.; Chang, I.; Lee, J.; Ryu, H. Non-Cell Autonomous and Epigenetic Mechanisms of Huntington’s Disease. Int. J. Mol. Sci. 2021, 22, 12499.
  116. Seol, Y.; Ki, S.; Ryu, H.L.; Chung, S.; Lee, J.; Ryu, H. How Microglia Manages Non-cell Autonomous Vicious Cycling of Aβ Toxicity in the Pathogenesis of AD. Front. Mol. Neurosci. 2020, 13, 593724.
  117. Lim, S.; Kim, H.-J.; Kim, D.-K.; Lee, S.-J. Non-cell-autonomous actions of α-synuclein: Implications in glial synucleinopathies. Prog. Neurobiol. 2018, 169, 158–171.
  118. Ilieva, H.; Polymenidou, M.; Cleveland, D.W. Non–cell autonomous toxicity in neurodegenerative disorders: ALS and beyond. J. Cell Biol. 2009, 187, 761–772.
  119. Vaz, S.H.; Pinto, S.; Sebastião, A.M.; Brites, D. Astrocytes in Amyotrophic Lateral Sclerosis; Araki, T., Ed.; Exon Publications: Brisbane, Australia, 2021; ISBN 978-0-6450017-7-8.
  120. McGeer, P.L.; McGeer, E.G. Inflammatory processes in amyotrophic lateral sclerosis. Muscle Nerve 2002, 26, 459–470.
  121. Beers, D.R.; Henkel, J.S.; Xiao, Q.; Zhao, W.; Wang, J.; Yen, A.A.; Siklos, L.; McKercher, S.R.; Appel, S.H. Wild-type microglia extend survival in PU.1 knockout mice with familial amyotrophic lateral sclerosis. Proc. Natl. Acad. Sci. USA 2006, 103, 16021–16026.
  122. Beers, D.R.; Henkel, J.S.; Zhao, W.; Wang, J.; Appel, S.H. CD4+ T cells support glial neuroprotection, slow disease progression, and modify glial morphology in an animal model of inherited ALS. Proc. Natl. Acad. Sci. USA 2008, 105, 15558–15563.
  123. Chiu, I.M.; Phatnani, H.; Kuligowski, M.; Tapia, J.C.; Carrasco, M.A.; Zhang, M.; Maniatis, T.; Carroll, M.C. Activation of innate and humoral immunity in the peripheral nervous system of ALS transgenic mice. Proc. Natl. Acad. Sci. USA 2009, 106, 20960–20965.
  124. Lincecum, J.M.; Vieira, F.G.; Wang, M.Z.; Thompson, K.; De Zutter, G.S.; Kidd, J.; Moreno, A.; Sanchez, R.; Carrion, I.J.; Levine, B.A.; et al. From transcriptome analysis to therapeutic anti-CD40L treatment in the SOD1 model of amyotrophic lateral sclerosis. Nat. Genet. 2010, 42, 392–399.
  125. Philips, T.; Rothstein, J.D. Glial cells in amyotrophic lateral sclerosis. Exp. Neurol. 2014, 262, 111–120.
  126. Zhao, W.; Beers, D.R.; Hooten, K.G.; Sieglaff, D.H.; Zhang, A.; Kalyana-Sundaram, S.; Traini, C.M.; Halsey, W.S.; Hughes, A.M.; Sathe, G.M.; et al. Characterization of Gene Expression Phenotype in Amyotrophic Lateral Sclerosis Monocytes Supplemental content. JAMA Neurol. 2017, 74, 677–685.
  127. Beers, D.R.; Appel, S.H. Immune dysregulation in amyotrophic lateral sclerosis: Mechanisms and emerging therapies. Lancet Neurol. 2019, 18, 211–220.
  128. Becher, B.; Spath, S.; Goverman, J. Cytokine networks in neuroinflammation. Nat. Rev. Immunol. 2017, 17, 49–59.
  129. Brites, D.; Vaz, A.R. Microglia centered pathogenesis in ALS: Insights in cell interconnectivity. Front. Cell. Neurosci. 2014, 8, 117.
  130. Gerber, Y.N.; Sabourin, J.C.; Hugnot, J.P.; Perrin, F.E. Unlike Physical Exercise, Modified Environment Increases the Lifespan of SOD1G93A Mice However Both Conditions Induce Cellular Changes. PLoS ONE 2012, 7, e45503.
  131. Alexianu, M.E.; Kozovska, M.; Appel, S.H. Immune reactivity in a mouse model of familial ALS correlates with disease progression. Neurology 2001, 57, 1282–1289.
  132. Chiu, I.M.; Morimoto, E.T.A.A.; Goodarzi, H.; Liao, J.T.; O’Keeffe, S.; Phatnani, H.P.; Muratet, M.; Carroll, M.C.; Levy, S.; Tavazoie, S.; et al. A neurodegeneration-specific gene-expression signature of acutely isolated microglia from an amyotrophic lateral sclerosis mouse model. Cell Rep. 2013, 4, 385–401.
  133. Geloso, M.C.; Corvino, V.; Marchese, E.; Serrano, A.; Michetti, F.; D’Ambrosi, N. The dual role of microglia in ALS: Mechanisms and therapeutic approaches. Front. Aging Neurosci. 2017, 9, 242.
  134. Tomiyama, M.; Kimura, T.; Maeda, T.; Tanaka, H.; Furusawa, K.; Kurahashi, K.; Matsunaga, M. Expression of metabotropic glutamate receptor mRNAs in the human spinal cord: Implications for selective vulnerability of spinal motor neurons in amyotrophic lateral sclerosis. J. Neurol. Sci. 2001, 189, 65–69.
  135. Aronica, E.; Catania, M.V.; Geurts, J.; Yankaya, B.; Troost, D. Immunohistochemical localization of group I and II metabotropic glutamate receptors in control and amyotrophic lateral sclerosis human spinal cord: Upregulation in reactive astrocytes. Neuroscience 2001, 105, 509–520.
  136. Valerio, A.; Ferrario, M.; Paterlini, M.; Liberini, P.; Moretto, G.; Cairns, N.J.; Pizzi, M.; Spano, P.F. Spinal cord mGlu1a receptors: Possible target for amyotrophic lateral sclerosis therapy. Pharmacol. Biochem. Behav. 2002, 73, 447–454.
  137. Anneser, J.M.H.; Ince, P.G.; Shaw, P.J.; Borasio, G.D. Differential expression of mGluR5 in human lumbosacral motoneurons. Neuroreport 2004, 15, 271–273.
  138. Anneser, J.M.H.; Chahli, C.; Borasio, G.D. Protective effect of metabotropic glutamate receptor inhibition on amyotrophic lateral sclerosis-cerebrospinal fluid toxicity in vitro. Neuroscience 2006, 141, 1879–1886.
  139. Ma, L.; Ostrovsky, H.; Miles, G.; Lipski, J.; Funk, G.D.; Nicholson, L.F.B. Differential expression of group I metabotropic glutamate receptors in human motoneurons at low and high risk of degeneration in amyotrophic lateral sclerosis. Neuroscience 2006, 143, 95–104.
  140. Anneser, J.M.H.; Chahli, C.; Ince, P.G.; Borasio, G.D.; Shaw, P.J. Glial proliferation and metabotropic glutamate receptor expression in amyotrophic lateral sclerosis. J. Neuropathol. Exp. Neurol. 2004, 63, 831–840.
  141. Planas-Fontánez, T.M.; Dreyfus, C.F.; Saitta, K.S. Reactive Astrocytes as Therapeutic Targets for Brain Degenerative Diseases: Roles Played by Metabotropic Glutamate Receptors. Neurochem. Res. 2020, 45, 541–550.
  142. Vergouts, M.; Doyen, P.J.; Peeters, M.; Opsomer, R.; Hermans, E. Constitutive downregulation protein kinase C epsilon in hSOD1(G93A) astrocytes influences mGluR5 signaling and the regulation of glutamate uptake. Glia 2018, 66, 749–761.
  143. Rossi, D.; Brambilla, L.; Valori, C.F.; Roncoroni, C.; Crugnola, A.; Yokota, T.; Bredesen, D.E.; Volterra, A. Focal degeneration of astrocytes in amyotrophic lateral sclerosis. Cell Death Differ. 2008, 15, 1691–1700.
  144. Vermeiren, C.; De Hemptinne, I.; Vanhoutte, N.; Tilleux, S.; Maloteaux, J.-M.M.; Hermans, E.; Hemptinne, I.; Vanhoutte, N.; Tilleux, S.; Maloteaux, J.-M.M.; et al. Loss of metabotropic glutamate receptor-mediated regulation of glutamate transport in chemically activated astrocytes in a rat model of amyotrophic lateral sclerosis. J. Neurochem. 2006, 96, 719–731.
  145. D’Antoni, S.; Berretta, A.; Seminara, G.; Longone, P.; Giuffrida-Stella, A.M.; Battaglia, G.; Sortino, M.A.; Nicoletti, F.; Catania, M. V A prolonged pharmacological blockade of type-5 metabotropic glutamate receptors protects cultured spinal cord motor neurons against excitotoxic death. Neurobiol. Dis. 2011, 42, 252–264.
  146. Martorana, F.; Brambilla, L.; Valori, C.F.; Bergamaschi, C.; Roncoroni, C.; Aronica, E.; Volterra, A.; Bezzi, P.; Rossi, D. The BH4 domain of Bcl-X(L) rescues astrocyte degeneration in amyotrophic lateral sclerosis by modulating intracellular calcium signals. Hum. Mol. Genet. 2012, 21, 826–840.
  147. Berger, J.V.; Dumont, A.O.; Focant, M.C.; Vergouts, M.; Sternotte, A.; Calas, A.-G.G.; Goursaud, S.; Hermans, E. Opposite regulation of metabotropic glutamate receptor 3 and metabotropic glutamate receptor 5 by inflammatory stimuli in cultured microglia and astrocytes. Neuroscience 2012, 205, 29–38.
  148. Bonifacino, T.; Musazzi, L.; Milanese, M.; Seguini, M.; Marte, A.; Gallia, E.; Cattaneo, L.; Onofri, F.; Popoli, M.; Bonanno, G. Altered mechanisms underlying the abnormal glutamate release in amyotrophic lateral sclerosis at a pre-symptomatic stage of the disease. Neurobiol. Dis. 2016, 95, 122–133.
  149. Milanese, M.; Zappettini, S.; Onofri, F.; Musazzi, L.; Tardito, D.; Bonifacino, T.; Messa, M.; Racagni, G.; Usai, C.; Benfenati, F.; et al. Abnormal exocytotic release of glutamate in a mouse model of amyotrophic lateral sclerosis. J. Neurochem. 2011, 116, 1028–1042.
  150. Milanese, M.; Zappettini, S.; Jacchetti, E.; Bonifacino, T.; Cervetto, C.; Usai, C.; Bonanno, G. In vitro activation of GAT1 transporters expressed in spinal cord gliosomes stimulates glutamate release that is abnormally elevated in the SOD1/G93A(+) mouse model of amyotrophic lateral sclerosis. J. Neurochem. 2010, 113, 489–501.
  151. Milanese, M.; Giribaldi, F.; Melone, M.; Bonifacino, T.; Musante, I.; Carminati, E.; Rossi, P.I.A.; Vergani, L.; Voci, A.; Conti, F.; et al. Knocking down metabotropic glutamate receptor 1 improves survival and disease progression in the SOD1(G93A) mouse model of amyotrophic lateral sclerosis. Neurobiol. Dis. 2014, 64, 48–59.
  152. Bonifacino, T.; Cattaneo, L.; Gallia, E.; Puliti, A.; Melone, M.; Provenzano, F.; Bossi, S.; Musante, I.; Usai, C.; Conti, F.; et al. In-vivo effects of knocking-down metabotropic glutamate receptor 5 in the SOD1(G93A) mouse model of amyotrophic lateral sclerosis. Neuropharmacology 2017, 123, 433–445.
  153. Bonifacino, T.; Provenzano, F.; Gallia, E.; Ravera, S.; Torazza, C.; Bossi, S.; Ferrando, S.; Puliti, A.; Van Den Bosch, L.; Bonanno, G.; et al. In-vivo genetic ablation of metabotropic glutamate receptor type 5 slows down disease progression in the SOD1(G93A) mouse model of amyotrophic lateral sclerosis. Neurobiol. Dis. 2019, 129, 79–92.
  154. Milanese, M.; Bonifacino, T.; Torazza, C.; Provenzano, F.; Kumar, M.; Ravera, S.; Arianna; Zerbo, R.; Frumento, G.; Balbi, M.; et al. Blocking glutamate mGlu 5 receptors with the negative allosteric modulator CTEP improves disease course in SOD1 G93A mouse model of amyotrophic lateral sclerosis. Br. J. Pharmacol. 2021, 178, 3747–3764.
More
This entry is offline, you can click here to edit this entry!
Video Production Service