Next Article in Journal
Mesenchymal Stem Cell Induced Foxp3(+) Tregs Suppress Effector T Cells and Protect against Retinal Ischemic Injury
Next Article in Special Issue
Differences in Immune-Related Genes Underlie Temporal and Regional Pathological Progression in 3xTg-AD Mice
Previous Article in Journal
Response to Electrostimulation Is Impaired in Muscle Cells from Patients with Chronic Obstructive Pulmonary Disease
Previous Article in Special Issue
Alzheimer’s Disease, Sleep Disordered Breathing, and Microglia: Puzzling out a Common Link
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dysfunction of RNA/RNA-Binding Proteins in ALS Astrocytes and Microglia

Istituto di Farmacologia Traslazionale, CNR, 00133 Rome, Italy
*
Authors to whom correspondence should be addressed.
Cells 2021, 10(11), 3005; https://doi.org/10.3390/cells10113005
Submission received: 28 September 2021 / Revised: 30 October 2021 / Accepted: 2 November 2021 / Published: 3 November 2021

Abstract

:
Amyotrophic Lateral Sclerosis is a neurological disease that primarily affects motor neurons in the cortex, brainstem, and spinal cord. The process that leads to motor neuron degeneration is strongly influenced by non-motor neuronal events that occur in a variety of cell types. Among these, neuroinflammatory processes mediated by activated astrocytes and microglia play a relevant role. In recent years, it has become clear that dysregulation of essential steps of RNA metabolism, as a consequence of alterations in RNA-binding proteins (RBPs), is a central event in the degeneration of motor neurons. Yet, a causal link between dysfunctional RNA metabolism and the neuroinflammatory processes mediated by astrocytes and microglia in ALS has been poorly defined. In this review, we will discuss the available evidence showing that RBPs and associated RNA processing are affected in ALS astrocytes and microglia, and the possible mechanisms involved in these events.

1. Introduction

Amyotrophic Lateral Sclerosis (ALS) is a disease of motor neurons and neighbouring non-neuronal cells. ALS affects humans either as a sporadic, non-familial form or as a less frequent familial form, with an autosomal dominant inheritance pattern [1]. In both cases, several associated/causative gene mutations have been identified, and thus the study of genetic ALS is at the centre of intense research efforts in devising effective therapeutic strategies that are still lacking [2,3].
The vast majority of genetic ALS is attributable to mutations in the SOD1, TDP-43, FUS, and C9orf72 genes [4]. The mechanisms which account for ALS have been deeply investigated in the last 30 years, and impairment in protein quality control, alterations in mitochondrial function, cytoskeletal and axonal defects, and dysfunctional RNA metabolism have emerged as leading pathogenic processes. Yet, many other mechanisms have been shown to play a role, further testifying for the complexity and the multifactorial nature of the disease [5]. Studies on SOD1, which was the first gene to be identified in familial ALS [6], established the paradigm that ALS is a non-cell-autonomous disease, with non-motor neuronal cells, such as astrocytes, microglia, muscle cells, oligodendrocytes, and others, being equally affected by the disease mechanisms and having an active role in the overall disease process. In particular, neuroinflammatory events orchestrated by activated astrocytes and microglia cells occur in both sporadic and familial ALS and play a crucial role in disease progression [7,8]. More recently, a contribution of the immune response in ALS progression has been established, thus adding further complexity to the multi-systemic nature of this disease [9,10]. Studies on FUS, TDP-43, and C9orf72 have introduced the concept that alterations of several processes that regulate RNA metabolism have a key role in pathogenic events. Indeed, RNA transcription, splicing, transport, stability, and translation, are affected in ALS motor neurons by both dysfunctional mutant TDP-43 and FUS, two RNA-binding proteins (RBPs) with established roles in the control of RNA metabolism, as well as by GGGGCC (G4C2) hexanucleotide repeat expansions (HRE) in the first intron of the C9orf72 gene, which affect RNA processing by an RNA-mediated gain-of-function mechanism [11,12]. Whether the two paradigms are connected, i.e., whether there is an interplay between defects in RBP/RNA metabolism and neuroinflammation in ALS, is still poorly characterized. In this review, we want to provide an overview of what has been described so far about a possible role of reactive microglia and astrocytes in ALS linked to TDP-43, FUS, and C9orf72 mutations, and how dysfunctional RBP and associated RNA processing might be involved in these events.

2. Glia-Mediated Neuroinflammation in TDP-43, FUS, and C9orf72 ALS

Astrocytes and microglia are essential cell types in the central nervous system (CNS), performing a vast range of functions, which are complementary to each other, to maintain the proper homeostasis of the CNS. Astrocytes, which are the most abundant non-neuronal cells in the CNS, provide metabolic support for neurons, are involved in the maintenance of blood–brain barrier integrity, modulate neuronal activity through the secretion of cytokines, chemokines, and gliotransmitters, and by controlling the proper ionic and neurotransmitter homeostasis, including glutamate buffering [13]. Microglia are the resident immune-competent cells of CNS, and constantly control the local microenvironment thus rapidly reacting to any stimuli that perturb the CNS homeostasis, such as infections or neuronal injury [14]. Further, microglia exert immune-independent functions in the developing as well as in the adult brain, thus extending the functional relevance of this cell type. In neurodegenerative conditions such as those characterizing ALS, microglia fine-tune their gene expression profiles and assume a range of different reactive states, which can be associated with different phases of the disease [15,16]. Similarly, astrocytes become reactive in response to various stimuli, a process commonly referred to as astrogliosis: they change their morphology, proliferate, and secrete pro- or anti-inflammatory molecules [17,18,19].
Neuroinflammation, characterized by a chronic activation of microglia and astrogliosis, is a pathological hallmark of ALS pathogenesis [20]. Neuroinflammation is indeed observed at sites of motor neuron degeneration in ALS patients, both in post-mortem tissues and in vivo during disease progression using PET studies [21,22,23,24,25,26]. Accordingly, neuroinflammation has been found in rodent models of SOD1-ALS (reviewed in [27]), as well as in murine models associated with mutations in TDP-43 [28,29,30,31,32,33], FUS [34,35,36,37,38], and C9orf72 [39,40,41,42,43,44]. Although microglia and astrocyte activation are supposed to be beneficial to neuronal homeostasis, a persistent reactive state can be detrimental, and a growing amount of evidence has demonstrated that glial cells actively contribute to the onset and progression of ALS. Several studies on SOD1-ALS, mostly performed in cells and mouse models, indeed indicate that motor neuron degeneration results from the combination of motor neuron cell-autonomous mechanisms and toxicity from neighbouring non-neuronal cells, including microglia and astrocytes, in a non-cell-autonomous manner [45]. Even though data are still limited, increasing evidence suggests that a role of non-cell-autonomous toxicity might be also extended to the other ALS gene mutations, as well as to the sporadic forms of the disease (Figure 1). Indeed, although in vitro studies show that astrocytes overexpressing an ALS-linked mutant (M337V) TDP-43 or lacking TDP-43 do not affect the survival of co-cultured wild-type motor neurons [46,47], a recent study demonstrates neuronal toxicity induced by co-cultured primary astrocytes overexpressing TDP-43 [48]. This is consistent with the in vivo observation that the selective expression of M337V-TDP-43 in rat astrocytes is sufficient to induce a progressive loss of motor neurons, denervation of skeletal muscles, and consequent paralysis [31]. Moreover, different reports demonstrate that motor neurons undergo cell death when co-cultured with fibroblast-derived astrocytes from sporadic or C9orf72-ALS patients [49,50,51,52,53]. Since control motor neurons survive as a monoculture in normal media, it is conceivable that motor neuron death is mediated by a gain of toxic function mechanism rather than a lack of trophic support from astrocytes [51]. Consistently, decreased neuronal cell viability can be caused by conditioned medium from cultured mouse and human astrocytes lacking TDP-43 expression [54], expressing mutant forms of TDP-43 [55], FUS [56], and C9orf72 [57], or even overexpressing wild-type FUS [58], which is known to cause ALS pathology in patients [59,60], and wild-type TDP-43 [48]. Altogether, these results suggest that astrocytes can extracellularly secrete factors that might be toxic to neurons, including proinflammatory cytokines such as IL-1β, IL-6, and tumour necrosis factor-α (TNFα) [48,56]. A role for TNFα and its receptors (TNFR1/TNFR2), as well as for other death receptor signalling, has been already implicated in ALS pathogenesis, with major findings coming from studies in SOD1-G93A mice [61]. However, an involvement of TNFα signalling has also been observed in TDP-43 pathology [62,63,64]. Even TDP-43 aggregates can propagate from human iPSC-derived astrocytes to motor neurons in co-culture experiments [65]. Yet, the opposite direction of spreading seems to be preferential, and this process might underlie a neuroprotective action of astrocytes, since they are able to reduce mislocalized cytoplasmic TDP-43, TDP-43 aggregation, and cell toxicity in co-cultured motor neurons [65].
Mechanisms underlying non-cell-autonomous motor neuron death are still unknown. Nevertheless, it has been observed that, when co-cultured with human C9orf72 iPSC-derived astrocytes, motor neurons undergo a progressive loss of action potential output, which can be reversed by CRISPR/Cas-9 mediated excision of the repeat expansion [53]. Moreover, C9orf72 ALS frontal cortex samples are characterized by decreased transcript levels of excitatory amino acid transporters (EAAT1 and EAAT2) compared to sporadic ALS [66]. EAAT1 and EAAT2 proteins are preferentially expressed in astrocytes [67] and are responsible for glutamate uptake from synapses [68]. Thus, their reduced expression leads to excitotoxicity and progressive paralysis in rodents [68]. Interestingly, decreased EAAT1 and EAAT2 expression has also been observed in the spinal cord of transgenic rats expressing astrocytic M337V-TDP-43 [31], as well as in Drosophila overexpressing TDP-43 homologous (TBPH) in glial cells [69]. Further, conditioned medium from mouse astrocytes expressing a mutant (R521G) FUS affects the expression levels of AMPA receptor subunits GluA1 and GluA2 in cultured motor neurons, leading, also in this case, to excitotoxic damage [56]. Finally, both the expression of the wild-type form or different mutant variants of TDP-43 and the reduction of TBPH in Drosophila glial cells result in alterations in neuromuscular junction morphology, associated with defective synaptic signals and locomotor dysfunction [48,70,71]. The deletion of astroglial TDP-43 in mice also induces motor dysfunction [72].

3. Dysfunction of RNA/RBPs in ALS Microglia and Astrocytes

In the context of the inflammatory events that have been highlighted in models of TDP-43, FUS, and C9orf72 ALS, an obvious question is as to whether evidence exists showing that dysfunctional TDP-43 and FUS induce glia-specific alterations that might underlie those events. Similarly, whether toxic RNAs and poly-dipeptide repeat (DPR) proteins originating from C9orf72 repeat expansion have a direct impact on glia is unclear. Although much further work will be needed to resolve the issue, several observations, that will be summarized in the following paragraphs, have started to provide answers to these questions.

3.1. TDP-43 and FUS

Mutations in TDP-43 and FUS have been associated with a significant fraction of both familial and sporadic ALS cases [73,74,75]. Both proteins are ubiquitously expressed DNA/RNA-binding proteins belonging to the family of heterogeneous nuclear ribonucleoproteins (hnRNPs). At a steady state, they localize predominantly in the nucleus, where they participate in different steps of gene expression regulation, including transcription, pre-mRNA splicing, and mRNA stability. However, they can shuttle continuously from nucleus to cytoplasm, where they are involved in other steps of RNA processing, including mRNA transport, translation and stress granule dynamics [76,77]. Most ALS mutations in both TDP-43 and FUS cause their re-localization from the nucleus to the cytoplasm, where they accumulate as protein aggregates. TDP-43 and FUS protein inclusions in affected neurons are indeed pathological hallmarks of ALS patients carrying respective mutations [73,78,79]. However, in 2006, wild-type TDP-43 was identified as the main component of ubiquitin-positive cytoplasmic aggregates of all cases of familial and sporadic ALS, with the notable exception of SOD1 and FUS patients, thus suggesting a more general role of TDP-43 in ALS pathology [80,81]. Pathogenic mechanisms whereby TDP-43 and FUS mutations cause ALS are still unclear. However, their subcellular mislocalization and aggregation suggest that both a loss of function due to their nuclear depletion and a gain of toxic function in the cytoplasm might contribute to ALS. According to the multiple functional roles of TDP-43 and FUS in RNA metabolism, alterations in different steps of this pathway might indeed have a primary role in motor neuron degeneration [11].
In addition to neuronal inclusions, glia-specific TDP-43 and FUS aggregates have been detected in ALS patients [25,73,78,79,80], thus suggesting that even both loss and gain-of-function mechanisms in microglia and astrocytes bearing mutant TDP-43 and FUS might be involved in the disease pathogenesis. Accordingly, glia-specific knockdown or overexpression of Drosophila TBPH (the fly homologue of mammalian TDP-43) are sufficient to cause, respectively, age-related motor deficits and premature lethality during larval stages [69,82]. Although the molecular mechanisms underlying TDP-43 and FUS toxicity in microglia and astrocytes are not yet defined, alterations in several cellular processes have been observed in different models in vitro and in vivo. It is known that in response to different environmental stressors both TDP-43 and FUS localize into cytoplasmic stress granules, transient membrane-less organelles where untranslated mRNAs, translational factors, and several RNA-binding proteins are stored during cell recovery, and proper dynamics of stress granules formation and dissolution is essential for cell survival. Khalfallah et al. have demonstrated that TDP-43 is critical for optimal stress granules dynamics both in mouse primary neurons and astrocytes exposed to oxidative stress [83]. Moreover, defects in glutathione induction and in the related antioxidant response following sodium arsenite treatment have been observed in primary astrocytes cultured from mutant Q331K-TDP-43 transgenic mice, as well as in M337V-TDP-43 patient fibroblasts [84]. Further, evidence for defects in astroglial metabolism has been recently provided. In particular, primary astrocytes derived from mutant transgenic mice (A315T-TDP-43) show dysfunctions in glutamate uptake and ATP accumulation [85], while the expression in rat cortical astrocytes of the C-terminal fragment of TDP-43 (208-414 aa), that forms cytoplasmic inclusions, affects lipid and glucose metabolism, thus reducing lactate release capacity, a crucial neuronal energy fuel [86].
Further, it has been observed that altered expression of TDP-43 and FUS in astrocytes and/or microglia is sufficient to modify their reactive states. In particular, mouse and human neural progenitor cells-derived astrocytes overexpressing wild-type FUS show increased reactivity to a proinflammatory stimulus (IL1β), compared to control astrocytes, and can induce microglia activation and neuronal cell death [58]. Similarly, the overexpression of TDP-43 in both primary microglial cells and astrocytes underlies hyperactive inflammatory response to lipopolysaccharide (LPS) or reactive oxygen species (ROS), with a higher production of proinflammatory cytokines and neurotoxic mediators compared to non-transgenic glial cells. Such response eventually increases microglia-mediated toxicity toward cultured cortical neurons, which can be reduced by inhibiting NF-kB activity [87]. Likewise, the loss of TDP-43 in primary cultured rodent microglia, but not in astrocytes, induces the expression of proinflammatory enzyme cyclooxygenase-2 (COX-2) through MAPK/ERK pathway activation, leading to the release of one of its key downstream products, prostaglandin E2 (PGE2), and eventually causing neuronal death [88]. Moreover, knockdown of TDP-43 in primary rat astrocytes causes impaired RNA homeostasis, as demonstrated by the accumulation of repetitive element transcripts and dsRNA, thus leading to a proinflammatory phenotype and to astrocyte activation [89].
Interestingly, not only endogenous levels of TDP-43 in microglia and astrocytes can modulate their activity, but it has been demonstrated that also aggregated species present in the extracellular milieu, which are found in biologic fluids of patients [90,91,92] and are considered to have prion-like properties (reviewed in [93]), can be internalized by cultured mouse microglia and induce an inflammatory response, mediated by caspase-3 activation and IL-1β and IL-18 signalling, which is cytotoxic for motor neuronal-like cultured cells [94]. Similar effects have been obtained by the exposure of mouse primary cultures of microglia to extracellular soluble forms of wild-type and mutant TDP-43 variants [64], further supporting the importance of non-cell-autonomous proinflammatory signalling in motor neuron injury caused by TDP-43.

3.2. C9orf72

How the hexanucleotide (G4C2) repeat expansion in the first intron of the C9orf72 gene might cause ALS neurodegeneration is currently unclear. However, both the loss of function of the protein encoded by the C9orf72 gene and a gain of function of RNA transcripts containing the expanded repeat have been to date hypothesized to mediate motor neuron degeneration [95].
C9orf72 protein acts in endosomal trafficking and autophagy pathway [96,97,98,99,100,101,102,103] and has a central role in maintaining immune homeostasis [104,105,106]. C9orf72 knockout mice indeed show alterations in the immune system with progressive splenomegaly, lymphadenopathy, and neuroinflammation, without showing any typical ALS neuropathological features and motor neuron degeneration [104,106]. Despite these findings support the hypothesis that gain-of-function mechanisms are primarily involved in C9orf72 disease, mouse models expressing C9orf72 repeat expansions exhibit varying degrees of neurodegeneration and do not completely recapitulate ALS pathology [40,41,107,108]. Further, haploinsufficiency in C9orf72 human motor neurons induces neurodegeneration [109]. Therefore, both loss and gain of function might contribute synergically to the disease onset and progression.
Although C9orf72 expression levels largely change across cell types and brain regions, the C9orf72 gene is highly expressed in microglia and, even if to a much lower extent, also in astrocytes [106,110], thus suggesting potentially pathological consequences of C9orf72 mutation on their proper homeostasis and functionality.
Reduced C9orf72 transcript and protein levels have been confirmed in different brain areas of post-mortem tissues of C9orf72-ALS patients (recently reviewed in [111]), but it is still unclear whether this also occurs specifically in microglia and astrocytes. However, it has been reported that in mice, the loss of C9orf72 protein induces the upregulation of proinflammatory cytokines, altered immune responses, and lysosome accumulation in microglia, thus potentially leading to defects in the ability to remove aggregated proteins [106]. Similarly, C9orf72 knockdown in U87 glioblastoma cells or normal human astrocytes induces the formation of p62 inclusions [66], a pathological feature of C9orf72-ALS [112]. Further, these cells show altered expression of thousands of genes, including genes involved in the immune system and glutamate-associated pathways, leading to defects in glutamate metabolism [66].
Sense and antisense RNA foci, generated by the intracellular accumulation of expanded RNA transcripts, represent specific pathological hallmarks of C9orf72-ALS neurons [113]. Nuclear RNA foci have been also found both in microglia and in astrocytes, even if to a lesser extent compared to neurons (reviewed in [114]). The lower prevalence might be explained by several reasons, including lower RNA expression or different RNA stability. Yet, this observation is a proof of concept that G4C2 expansion might induce toxicity in these cells. In neurons, RNA foci are suggested to cause defects in RNA processing through sequestration of RNA-binding proteins [115,116,117,118,119,120,121], but it is still little explored if this might also occur in glial cells. However, a first clue comes from a report that shows a significant colocalization of the splicing factor hnRNP H with G4C2 RNA foci in cultured astrocytes from C9orf72 post-mortem tissues compared to control cells [122]. Importantly, hnRNP H sequestration has also been observed in post-mortem tissues from C9orf72-ALS patients [115,122,123], and consequent aberrant alternative splicing of multiple known hnRNP H targets has been detected in brains from both C9orf72 and sporadic ALS patients [122,124,125]. Altogether, these findings support a crucial role of hnRNP H in ALS pathogenesis, and further studies are necessary to better understand the functional relevance of the interaction between hnRNP H (as well as other RBPs) and expanded G4C2 transcripts in glial cells.
Another specific pathological hallmark of C9orf72-ALS neurons is the intracellular accumulation of five poly-dipeptide repeat (DPR) proteins (poly-GA, -GP, -GR, -PR, and -PA), unconventionally produced by repeat-associated non-AUG (RAN) translation from sense and antisense expanded transcripts [126]. In neurons, DPR toxicity seems to be mediated by the aberrant interactions of DPRs with several proteins, thus affecting their normal cellular function and leading to defects in different molecular pathways, including RNA processing and nucleus-cytoplasmic transport [127]. Despite the fact that no DPR protein inclusions are detected in glial cells both in subcortical white matter, hippocampus and white matter in the spinal cord of C9orf72-ALS post-mortem tissues [128], their expression cannot be completely excluded in these cell types. Indeed, the presence of soluble forms of DPR proteins might still compromise cell functionality, independently from their accumulation into detectable inclusions. Interestingly, depletion of the nuclear export adaptor SRSF1 in C9orf72 patient-derived astrocytes prevents motor neuron death in co-culture assays [117]. SRSF1 mediates, in concert with NXF1, the nuclear export of expanded C9orf72 RNA transcripts and its depletion are able to reduce cytoplasmic DPR expression thus preventing neurodegeneration in both in vitro and in vivo models [117]. Thus, it is conceivable that DPR expression might underlie the observed astrocyte-induced neurotoxicity.
TDP-43-positive inclusions, which characterize C9orf72-ALS, as well as other sporadic and familial cases, have been reported in glia in different brain regions, as well as in the spinal cord of C9orf72-ALS cases [21,112,129]. These findings thus reinforce the hypothesis that alterations in RNA metabolism might occur in glial cells and contribute to ALS pathogenesis, although further investigations will be needed to understand the underlying mechanisms.
The precise molecular cascade induced by C9orf72 mutation within microglia and astrocytes is still unknown. Yet, increasing data imply mechanisms of cell-autonomous toxicity in glial cells. It has been found that reactive microglia of C9orf72-ALS patient post-mortem motor cortex and spinal cord is characterized by a strong accumulation of LAMP1-positive lysosomes, differently from sporadic ALS samples [106], thus suggesting potential lysosomal alterations in C9orf72 cases, a pathway already strongly implicated in ALS pathology by genetic and experimental findings [130]. Altered levels of p62 and LAMP2A have been also found in microglial BV-2 cells expressing pathological repeat expansion, although functional phagocytic capability, appropriate response to an inflammatory stimulus, and normal cell viability were observed [114]. These results thus suggest that the repeat expansion does not affect microglia functional activity, but further studies in other models are needed to better understand the effect of mutation on microglial phenotypes.
Finally, both in astrocytes derived from sporadic ALS patients and in astrocytes from familial C9orf72 cases, defects in energy production have been described [131,132]. In particular, dysfunctions in adenosine, glycogen, and fructose metabolism, caused by reduced expression levels of key pathway enzymes, as well as defects in the membrane transport of mitochondrial energy substrates have been identified [131,132]. Such metabolic defects might result in insufficient nutritional support for neurons, thus potentially mediating neurotoxicity and contributing to disease progression. Indeed, circumventing adenosine metabolism dysfunction by inosine supplementation in vitro was able to improve the survival of motor neurons co-cultured with C9orf72 patient-derived astrocytes [131]. Additionally, accelerated senescence and defects in the production and/or secretion of antioxidants have been observed in human iPSC-derived astrocytes from C9orf72-ALS patients, thus leading to increased oxidative stress in astrocytes, as well as in co-cultured motor neurons, possibly contributing to neurotoxicity [49].

4. The Contribution of RNA Dysmetabolism to Neuroinflammation in ALS

As already mentioned, alterations in essential steps of RNA metabolism are currently believed to play key roles in motor neuron degeneration in ALS. Indeed, a wealth of experimental data demonstrated that defects in mRNA transcription and splicing, transport, and translations are associated with, and often causally involved in, the final degeneration of motor neurons (recently reviewed in [12,133]). While a specific step of RNA metabolism has not yet emerged as to be more relevant than the others, defects in mRNA splicing, transport and translation seem to play a major pathological role. Whether alterations in the latter processes are causally linked to the neuroinflammatory events that mark ALS progression is much less characterized. In the following paragraphs, we will therefore highlight the experimental evidence supporting this conclusion, as well as the circumstantial evidence that suggests that a link might indeed occur.

4.1. Alternative Splicing in Reactive ALS Glia

Alternative splicing is a key process of gene expression regulation, so it is not surprising that in ALS pathological conditions that are characterized by profound changes in gene expression, alterations in underlying splicing regulation do play a role. A wealth of data has been obtained from ALS motor neurons derived from rodent models of the disease as well as from patient-derived cells, showing that defective splicing regulation occurs in ALS [36,122,125,134,135,136,137,138,139,140,141,142,143]. Much less is known about splicing dysregulation in ALS astrocytes and microglia. Early studies have provided support to the notion that alternative splicing dysregulation might also occur in astrocytes in ALS conditions. Indeed, cultured human astrocytes exposed to synthetic poly-GR and -PR C9orf72-derived dipeptides undergo significant changes in the alternative splicing of a number of mRNAs, including the mRNA encoding the glutamate transporter EAAT2, whose splicing is known to be affected in ALS patients [144,145]. This effect is possibly mediated by the interaction of C9orf72 DPRs with hnRNPA2, an RBP with known function in RNA splicing regulation, suggesting that DPR might impact on the activity of RBPs thereby affecting astrocyte functions. It is not clear, however, if these events occur in vivo in ALS astrocytes, where deposition of DPR protein inclusions are not easily detectable, as mentioned.
In genetic ALS cases due to mutations in RBPs that have a direct and established role on RNA splicing regulation, such as FUS and TDP-43, a number of potential mechanisms have been described to induce neurodegeneration. These include loss-of-function-dependent dysregulation of RNA splicing, as well as gain of splicing functions that lead to the aberrant inclusion of cryptic exons and/or introns into a range of mRNAs, including transcripts encoding RBPs [136,146], thus supporting the existence of a pathological vicious cycle that involves RBPs and that boosts splicing alterations in ALS motor neurons [147]. Similarly, dysregulation by C9orf72 repeat expansions in hnRNP H, another RBP with established roles in splicing regulation, is associated with a widespread intron retention program in human brains [141].
Recently, aberrant alternative splicing has been convincingly proposed to have a role in astrocyte reactivity in ALS. Valosin-containing protein (VCP), an ATPase involved in autophagosomes maturation, is mutated in 1–2% of familial ALS cases [148]. Using iPSC-derived astrocytes from ALS patients carrying the ALS-causing R155C and R191Q mutant VCP, Ziff et al. found that intron retention, which characterizes RNA transcripts from healthy astrocytes, is strikingly decreased [149]. This leads to a significant increase in the expression of the corresponding genes, which are particularly enriched into functional categories related to astrocyte reactivity regulation. Similar results were reproduced when analyzing human iPSC astrocytes derived from mutant SOD1 and C9orf72 ALS patients, as well as from astrocytes stimulated with TNFα, IL-1β, and C1q, indicating that diminished intron retention is a general feature of reactive astrocytes, including those generated by ALS pathogenic mutant genes. The mechanisms whereby VCP, SOD1, or C9orf72 mutations converge into regulation of intron retention of overlapping genes are so far unknown, but the observation from the same authors that astrocytes knocked-out for TDP-43 display a similar pattern of splicing alteration suggests that TDP-43 dysfunction, which is common to the majority of ALS cases, might have a role in the process. Importantly, these findings apparently diverge from what has been reported to occur in human motor neurons, where a significant increase in intron retention, coupled with a decreased stability and expression of the corresponding mRNAs, has been described [141,146,150,151]. The reasons for these differences are far from being clear, but they obviously indicate that the outcomes of RNA dysfunctions in ALS pathogenesis are likely cell-type specific and that therefore additional studies on RBP/RNA alterations in ALS astrocytes cells are urgently required to complete the picture of dysfunctional RNA metabolism in ALS pathogenesis. Similarly, whether and how alternative splicing alterations by dysfunctional RBPs might contribute to the detrimental effect of reactive microglia in ALS needs to be defined. Yet, it has been recently shown that misregulated alternative splicing in RBP-defective mouse microglia leads to the activation of the RhoA GTPase/ROCK pathways that, in turn, induce proinflammatory phenotypes. Importantly, these changes might be linked to several diseases of the nervous system, including neurodegenerative diseases, further supporting a possible role of these alterations in ALS [152].

4.2. RNA Trafficking and (Local) Translation in Reactive ALS Glia

Defective mRNA transport and translation are deeply implicated in ALS pathogenesis associated with FUS, TDP-43, and C9orf72 gene mutations. Indeed, both TDP-43 and FUS associate with proteins involved in mRNA transport, and are found in dendritic and axon terminals, supporting a role of both proteins in the regulation of mRNA transport in neurons. Moreover, they both regulate mRNA translation, either by binding to ribosomal subunits and translation factors and/or by regulating the availability of specific mRNAs for translation [76,153,154]. According to these functions, mutations in FUS and TDP-43 impact both the overall rate of translation efficiency, as well as on the synthesis of specific substrates (recently reviewed in [76]). Similarly, C9orf72 mutations elicit global protein synthesis repression by the sequestration of translation factors into G4C2 RNA foci or by interfering with translation through G4C2-derived DPRs [153,155,156,157,158]. This seems to also occur in human astrocytes, where poly-GR an -PR dipeptides induce significant changes in the expression of ribosomal rRNA genes and/or in the processing of rRNAs [144]. Moreover, it has been shown that poly-PR is able to decrease the rate of protein translation in mouse primary astrocytes [159], thus fostering the idea that C9orf72 might have a widespread impact on global translation efficiency. Notably, recent papers have revealed that ALS motor neuron pathology due to FUS mutations might be tightly linked to alterations in local translation regulation. It has been demonstrated, in fact, that ALS-linked mutant FUS accumulate into both cultured hippocampal neuron axons and sciatic nerve motor axons and that this is associated with a decreased axonal protein synthesis rate [160]. This suggests that FUS might have a role in synaptic local translation regulation, a hypothesis that is supported by the observation that in mouse and human models of FUS-ALS, FUS affects the localization and function of FMRP, another RBP with an established role in local translational regulation [161,162,163]. Importantly, glutamate receptor GLT1 expression in astrocytes and the consequent clearance of extracellular glutamate is diminished in mice knockout for FMRP [164]. This is due to a decreased expression of astroglial Gq-coupled metabotropic glutamate receptor mGluR5, whose mRNA is normally bound by FMRP and whose translation is impaired by the loss of FMRP. Although this is in contrast with the established role of FMRP as a translation inhibitor, these findings suggest the possibility, that needs to be demonstrated, that even in astrocytes an altered FMRP signalling by mutant ALS RBPs might affect local protein translation, which indeed occurs in the distal, astrocyte peri-synaptic processes [165].

5. Conclusions

The concept that dysregulation of the RNA-binding protein regulatory network, which controls all the aspects of RNA processing, is deeply involved in the degeneration of motor neurons in ALS has solid grounds in an impressive set of experimental data that have been produced in the very last years. This has been mostly obtained by studying motor neurons, which indeed are the key players in ALS, but the pathological features that typify ALS-linked mutant TDP-43, FUS, and C9orf72 in neuronal cells, seem to be reproduced, at least in part, in astrocytes and microglia cells, suggesting that RNA dysmetabolism might participate to the inflammatory processes that both cell types orchestrate during disease progression (Figure 2 and Figure 3).
Certainly, a better comprehension of the mechanisms whereby dysfunctional RBPs impact the different aspects of RNA metabolism regulation, and how this contributes to the noxious effects that reactive glia exerts on motor neurons will provide further support to this conclusion and novel pathological targets to be exploited in the search of therapeutic approaches for ALS.

Author Contributions

Conceptualization, S.R. and M.C.; writing—original draft preparation, S.R. and M.C.; writing—review and editing, S.R. and M.C.; visualization, S.R. and M.C.; supervision, S.R., funding acquisition, M.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Fondazione Italiana di Ricerca per la Sclerosi Laterale Amiotrofica (ARISLA), grant “SPLICEALS”.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Masrori, P.; van Damme, P. Amyotrophic lateral sclerosis: A clinical review. Eur. J. Neurol. 2020, 27, 1918–1929. [Google Scholar] [CrossRef] [PubMed]
  2. Van Es, M.; Hardiman, O.; Chio, A.; Al-Chalabi, A.; Pasterkamp, R.J.; Veldink, J.H.; van den Berg, L.H. Amyotrophic lateral sclerosis. Lancet 2017, 390, 2084–2098. [Google Scholar] [CrossRef]
  3. Brown, R.H., Jr.; Al-Chalabi, A. Amyotrophic lateral sclerosis. N. Engl. J. Med. 2017, 377, 1602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Kim, G.; Gautier, O.; Tassoni-Tsuchida, E.; Ma, X.R.; Gitler, A.D. ALS genetics: Gains, losses, and implications for future therapies. Neuron 2020, 108, 822–842. [Google Scholar] [CrossRef]
  5. Mejzini, R.; Flynn, L.; Pitout, I.L.; Fletcher, S.; Wilton, S.; Akkari, P.A. ALS genetics, mechanisms, and therapeutics: Where are we now? Front. Neurosci. 2019, 13, 1310. [Google Scholar] [CrossRef] [Green Version]
  6. Rosen, D.R.; Siddique, T.; Patterson, D.; Figlewicz, D.A.; Sapp, P.C.; Hentati, A.; Donaldson, D.H.; Goto, J.; O’Regan, J.P.; Deng, H.-X.; et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 1993, 362, 59–62. [Google Scholar] [CrossRef]
  7. Ilieva, H.; Polymenidou, M.; Cleveland, D.W. Non–cell autonomous toxicity in neurodegenerative disorders: ALS and beyond. J. Cell Biol. 2009, 187, 761–772. [Google Scholar] [CrossRef] [Green Version]
  8. Chen, H.; Kankel, M.W.; Su, S.C.; Han, S.W.S.; Ofengeim, D. Exploring the genetics and non-cell autonomous mechanisms underlying ALS/FTLD. Cell Death Differ. 2018, 25, 648–662. [Google Scholar] [CrossRef] [Green Version]
  9. Garofalo, S.; Cocozza, G.; Porzia, A.; Inghilleri, M.; Raspa, M.; Scavizzi, F.; Aronica, E.; Bernardini, G.; Peng, L.; Ransohoff, R.M.; et al. Natural killer cells modulate motor neuron-immune cell cross talk in models of amyotrophic lateral sclerosis. Nat. Commun. 2020, 11, 1–16. [Google Scholar] [CrossRef] [Green Version]
  10. Coque, E.; Salsac, C.; Espinosa-Carrasco, G.; Varga, B.; Degauque, N.; Cadoux, M.; Crabé, R.; Virenque, A.; Soulard, C.; Fierle, J.K.; et al. Cytotoxic CD8+ T lymphocytes expressing ALS-causing SOD1 mutant selectively trigger death of spinal motoneurons. Proc. Natl. Acad. Sci. USA 2019, 116, 2312–2317. [Google Scholar] [CrossRef] [Green Version]
  11. Butti, Z.; Patten, S.A. RNA dysregulation in amyotrophic lateral sclerosis. Front. Genet. 2019, 9, 712. [Google Scholar] [CrossRef] [Green Version]
  12. Nussbacher, J.K.; Tabet, R.; Yeo, G.W.; Lagier-Tourenne, C. Disruption of RNA metabolism in neurological diseases and emerging therapeutic interventions. Neuron 2019, 102, 294–320. [Google Scholar] [CrossRef] [PubMed]
  13. Vasile, F.; Dossi, E.; Rouach, N. Human astrocytes: Structure and functions in the healthy brain. Brain Struct. Funct. 2017, 222, 2017–2029. [Google Scholar] [CrossRef] [Green Version]
  14. Salter, M.W.; Stevens, B. Microglia emerge as central players in brain disease. Nat. Med. 2017, 23, 1018–1027. [Google Scholar] [CrossRef] [PubMed]
  15. Ransohoff, R.M. A polarizing question: Do M1 and M2 microglia exist? Nat. Neurosci. 2016, 19, 987–991. [Google Scholar] [CrossRef] [PubMed]
  16. Clarke, B.E.; Patani, R. The microglial component of amyotrophic lateral sclerosis. Brain 2020, 143, 3526–3539. [Google Scholar] [CrossRef] [PubMed]
  17. Liddelow, S.A.; Barres, B.A. Reactive astrocytes: Production, function, and therapeutic potential. Immunity 2017, 46, 957–967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.S.; Peterson, T.C.; et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef]
  19. Valori, C.F.; Guidotti, G.; Brambilla, L.; Rossi, D. Astrocytes: Emerging therapeutic targets in neurological disorders. Trends Mol. Med. 2019, 25, 750–759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. McCauley, M.E.; Baloh, R.H. Inflammation in ALS/FTD pathogenesis. Acta Neuropathol. 2019, 137, 715–730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Cooper-Knock, J.; Hewitt, C.; Highley, R.; Brockington, A.; Milano, A.; Man, S.; Martindale, J.; Hartley, J.; Walsh, T.; Gelsthorpe, C.; et al. Clinico-pathological features in amyotrophic lateral sclerosis with expansions in C9ORF72. Brain 2012, 135, 751–764. [Google Scholar] [CrossRef] [PubMed]
  22. Corcia, P.; Tauber, C.; Vercoullie, J.; Arlicot, N.; Prunier, C.; Praline, J.; Nicolas, G.; Venel, Y.; Hommet, C.; Baulieu, J.-L.; et al. Molecular imaging of microglial activation in amyotrophic lateral sclerosis. PLoS ONE 2012, 7, e52941. [Google Scholar] [CrossRef]
  23. Engelhardt, J.I.; Appel, S.H. IgG reactivity in the spinal cord and motor cortex in amyotrophic lateral sclerosis. Arch. Neurol. 1990, 47, 1210–1216. [Google Scholar] [CrossRef]
  24. Henkel, J.S.; Engelhardt, J.I.; Siklós, L.; Simpson, E.P.; Kim, S.H.; Pan, T.; Goodman, J.C.; Siddique, T.; Beers, D.R.; Appel, S.H. Presence of dendritic cells, MCP-1, and activated microglia/macrophages in amyotrophic lateral sclerosis spinal cord tissue. Ann. Neurol. 2003, 55, 221–235. [Google Scholar] [CrossRef]
  25. Nolan, M.; Scott, C.; Gamarallage, M.P.; Lunn, D.; Carpenter, K.; McDonough, E.; Meyer, D.; Kaanumalle, S.; Santamaria-Pang, A.; Turner, M.R.; et al. Quantitative patterns of motor cortex proteinopathy across ALS genotypes. Acta Neuropathol. Commun. 2020, 8, 1–20. [Google Scholar] [CrossRef] [PubMed]
  26. Turner, M.R.; Cagnin, A.; Turkheimer, F.E.; Miller, C.C.J.; Shaw, C.E.; Brooks, D.J.; Leigh, P.N.; Banati, R.B. Evidence of widespread cerebral microglial activation in amyotrophic lateral sclerosis: An [11C](R)-PK11195 positron emission tomography study. Neurobiol. Dis. 2004, 15, 601–609. [Google Scholar] [CrossRef]
  27. Philips, T.; Rothstein, J. Glial cells in amyotrophic lateral sclerosis. Exp. Neurol. 2014, 262, 111–120. [Google Scholar] [CrossRef] [Green Version]
  28. Hunter, M.; Spiller, K.J.; Dominique, M.A.; Xu, H.; Hunter, F.W.; Fang, T.C.; Canter, R.G.; Roberts, C.J.; Ransohoff, R.M.; Trojanowski, J.Q.; et al. Microglial transcriptome analysis in the rNLS8 mouse model of TDP-43 proteinopathy reveals discrete expression profiles associated with neurodegenerative progression and recovery. Acta Neuropathol. Commun. 2021, 9, 1–19. [Google Scholar] [CrossRef] [PubMed]
  29. Jara, J.H.; Gautam, M.; Kocak, N.; Xie, E.F.; Mao, Q.; Bigio, E.H.; Özdinler, P.H. MCP1-CCR2 and neuroinflammation in the ALS motor cortex with TDP-43 pathology. J. Neuroinflamm. 2019, 16, 196. [Google Scholar] [CrossRef]
  30. Swarup, V.; Phaneuf, D.; Bareil, C.; Robertson, J.; Rouleau, G.A.; Kriz, J.; Julien, J.-P. Pathological hallmarks of amyotrophic lateral sclerosis/frontotemporal lobar degeneration in transgenic mice produced with TDP-43 genomic fragments. Brain 2011, 134, 2610–2626. [Google Scholar] [CrossRef]
  31. Tong, J.; Huang, C.; Bi, F.; Wu, Q.; Huang, B.; Liu, X.; Li, F.; Zhou, H.; Xia, X.-G. Expression of ALS-linked TDP-43 mutant in astrocytes causes non-cell-autonomous motor neuron death in rats. EMBO J. 2013, 32, 1917–1926. [Google Scholar] [CrossRef]
  32. Wegorzewska, I.; Bell, S.; Cairns, N.J.; Miller, T.M.; Baloh, R.H. TDP-43 mutant transgenic mice develop features of ALS and frontotemporal lobar degeneration. Proc. Natl. Acad. Sci. USA 2009, 106, 18809–18814. [Google Scholar] [CrossRef] [Green Version]
  33. Xu, Y.-F.; Zhang, Y.-J.; Lin, W.-L.; Cao, X.; Stetler, C.; Dickson, D.W.; Lewis, J.; Petrucelli, L. Expression of mutant TDP-43 induces neuronal dysfunction in transgenic mice. Mol. Neurodegener. 2011, 6, 73. [Google Scholar] [CrossRef] [Green Version]
  34. Huang, C.; Tong, J.; Bi, F.; Wu, Q.; Huang, B.; Zhou, H.; Xia, X.-G. Entorhinal cortical neurons are the primary targets of FUS mislocalization and ubiquitin aggregation in FUS transgenic rats. Hum. Mol. Genet. 2012, 21, 4602–4614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Huang, C.; Zhou, H.; Tong, J.; Chen, H.; Liu, Y.-J.; Wang, D.; Wei, X.; Xia, X.-G. FUS transgenic rats develop the phenotypes of amyotrophic lateral sclerosis and frontotemporal lobar degeneration. PLoS Genet. 2011, 7, e1002011. [Google Scholar] [CrossRef] [Green Version]
  36. Mirra, A.; Rossi, S.; Scaricamazza, S.; di Salvio, M.; Salvatori, I.; Valle, C.; Rusmini, P.; Poletti, A.; Cestra, G.; Carri, M.T.; et al. Functional interaction between FUS and SMN underlies SMA-like splicing changes in wild-type hFUS mice. Sci. Rep. 2017, 7, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Mitchell, J.C.; McGoldrick, P.; Vance, C.; Hortobagyi, T.; Sreedharan, J.; Rogelj, B.; Tudor, E.L.; Smith, B.N.; Klasen, C.; Miller, C.C.; et al. Overexpression of human wild-type FUS causes progressive motor neuron degeneration in an age- and dose-dependent fashion. Acta Neuropathol. 2013, 125, 273–288. [Google Scholar] [CrossRef] [Green Version]
  38. Sharma, A.; Lyashchenko, A.K.; Lu, L.; Nasrabady, S.E.; Elmaleh, M.; Mendelsohn, M.; Nemes, A.; Tapia, J.C.; Mentis, G.Z.; Shneider, N.A. ALS-associated mutant FUS induces selective motor neuron degeneration through toxic gain of function. Nat. Commun. 2016, 7, 10465. [Google Scholar] [CrossRef]
  39. Brettschneider, J.; van Deerlin, V.M.; Robinson, J.L.; Kwong, L.; Lee, E.B.; Ali, Y.O.; Safren, N.; Monteiro, M.J.; Toledo, J.; Elman, L.; et al. Pattern of ubiquilin pathology in ALS and FTLD indicates presence of C9ORF72 hexanucleotide expansion. Acta Neuropathol. 2012, 123, 825–839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Chew, J.; Gendron, T.F.; Prudencio, M.; Sasaguri, H.; Zhang, Y.-J.; Castanedes-Casey, M.; Lee, C.W.; Jansen-West, K.; Kurti, A.; Murray, M.E.; et al. C9ORF72 repeat expansions in mice cause TDP-43 pathology, neuronal loss, and behavioral deficits. Science 2015, 348, 1151–1154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Liu, Y.; Pattamatta, A.; Zu, T.; Reid, T.; Bardhi, O.; Borchelt, D.R.; Yachnis, A.T.; Ranum, L.P.W. C9orf72 BAC mouse model with motor deficits and neurodegenerative features of ALS/FTD. Neuron 2016, 90, 521–534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Schludi, M.H.; Becker, L.; Garrett, L.; Gendron, T.F.; Zhou, Q.; Schreiber, F.; Popper, B.; Dimou, L.; Strom, T.M.; Winkelmann, J.; et al. Spinal poly-GA inclusions in a C9orf72 mouse model trigger motor deficits and inflammation without neuron loss. Acta Neuropathol. 2017, 134, 241–254. [Google Scholar] [CrossRef] [PubMed]
  43. Zhang, Y.-J.; Gendron, T.F.; Ebbert, M.T.W.; O’Raw, A.; Yue, M.; Jansen-West, K.; Zhang, X.; Prudencio, M.; Chew, J.; Cook, C.N.; et al. Poly (GR) impairs protein translation and stress granule dynamics in C9orf72-associated frontotemporal dementia and amyotrophic lateral sclerosis. Nat. Med. 2018, 24, 1136–1142. [Google Scholar] [CrossRef]
  44. LaClair, K.D.; Zhou, Q.; Michaelsen, M.; Wefers, B.; Brill, M.S.; Janjic, A.; Rathkolb, B.; Farny, D.; Cygan, M.; de Angelis, M.H.; et al. Congenic expression of poly-GA but not poly-PR in mice triggers selective neuron loss and interferon responses found in C9orf72 ALS. Acta Neuropathol. 2020, 140, 121–142. [Google Scholar] [CrossRef] [PubMed]
  45. Lee, J.; Hyeon, S.J.; Im, H.; Ryu, H.; Kim, Y.; Ryu, H. Astrocytes and microglia as non-cell autonomous players in the pathogenesis of ALS. Exp. Neurobiol. 2016, 25, 233–240. [Google Scholar] [CrossRef]
  46. Haidet-Phillips, A.M.; Gross, S.K.; Williams, T.; Tuteja, A.; Sherman, A.; Ko, M.; Jeong, Y.H.; Wong, P.C.; Maragakis, N.J. Altered astrocytic expression of TDP-43 does not influence motor neuron survival. Exp. Neurol. 2013, 250, 250–259. [Google Scholar] [CrossRef] [PubMed]
  47. Serio, A.; Bilican, B.; Barmada, S.J.; Ando, D.M.; Zhao, C.; Siller, R.; Burr, K.; Haghi, G.; Story, D.; Nishimura, A.L.; et al. Astrocyte pathology and the absence of non-cell autonomy in an induced pluripotent stem cell model of TDP-43 proteinopathy. Proc. Natl. Acad. Sci. USA 2013, 110, 4697–4702. [Google Scholar] [CrossRef] [Green Version]
  48. Lee, S.; Kim, S.; Kang, H.-Y.; Lim, H.R.; Kwon, Y.; Jo, M.; Jeon, Y.-M.; Kim, S.R.; Kim, K.; Ha, C.M.; et al. The overexpression of TDP-43 in astrocytes causes neurodegeneration via a PTP1B-mediated inflammatory response. J. Neuroinflamm. 2020, 17, 1–22. [Google Scholar] [CrossRef]
  49. Birger, A.; Ben-Dor, I.; Ottolenghi, M.; Turetsky, T.; Gil, Y.; Sweetat, S.; Perez, L.; Belzer, V.; Casden, N.; Steiner, D.; et al. Human iPSC-derived astrocytes from ALS patients with mutated C9ORF72 show increased oxidative stress and neurotoxicity. EBioMedicine 2019, 50, 274–289. [Google Scholar] [CrossRef] [Green Version]
  50. Haidet-Phillips, A.M.; Hester, M.E.; Miranda, C.; Meyer, K.; Braun, L.; Frakes, A.; Song, S.; Likhite, S.; Murtha, M.J.; Foust, K.D.; et al. Astrocytes from familial and sporadic ALS patients are toxic to motor neurons. Nat. Biotechnol. 2011, 29, 824–828. [Google Scholar] [CrossRef] [Green Version]
  51. Meyer, K.; Ferraiuolo, L.; Miranda, C.J.; Likhite, S.; McElroy, S.; Renusch, S.; Ditsworth, D.; Lagier-Tourenne, C.; Smith, R.A.; Ravits, J.; et al. Direct conversion of patient fibroblasts demonstrates non-cell autonomous toxicity of astrocytes to motor neurons in familial and sporadic ALS. Proc. Natl. Acad. Sci. USA 2014, 111, 829–832. [Google Scholar] [CrossRef] [Green Version]
  52. Qian, K.; Huang, H.; Peterson, A.; Hu, B.; Maragakis, N.J.; Ming, G.-L.; Chen, H.; Zhang, S.-C. Sporadic ALS astrocytes induce neuronal degeneration in vivo. Stem Cell Rep. 2017, 8, 843–855. [Google Scholar] [CrossRef] [PubMed]
  53. Zhao, C.; Devlin, A.; Chouhan, A.K.; Selvaraj, B.T.; Stavrou, M.; Burr, K.; Brivio, V.; He, X.; Mehta, A.R.; Story, D.; et al. Mutant C9orf72 human iPSC-derived astrocytes cause non-cell autonomous motor neuron pathophysiology. Glia 2019, 68, 1046–1064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Lu, L.; Zheng, L.; Si, Y.; Luo, W.; Dujardin, G.; Kwan, T.; Potochick, N.R.; Thompson, S.; Schneider, D.; King, P.H. Hu antigen R (HuR) is a positive regulator of the RNA-binding proteins TDP-43 and FUS/TLS. J. Biol. Chem. 2014, 289, 31792–31804. [Google Scholar] [CrossRef] [Green Version]
  55. Rojas, F.; Cortes, N.; Abarzua, S.; Dyrda, A.; van Zundert, B.A. Astrocytes expressing mutant SOD1 and TDP43 trigger motoneuron death that is mediated via sodium channels and nitroxidative stress. Front. Cell. Neurosci. 2014, 8, 24. [Google Scholar] [CrossRef] [Green Version]
  56. Kia, A.; McAvoy, K.; Krishnamurthy, K.; Trotti, D.; Pasinelli, P. Astrocytes expressing ALS-linked mutant FUS induce motor neuron death through release of tumor necrosis factor-alpha. Glia 2018, 66, 1016–1033. [Google Scholar] [CrossRef]
  57. Madill, M.; McDonagh, K.; Ma, J.; Vajda, A.; McLoughlin, P.; O’Brien, T.; Hardiman, O.; Shen, S. Amyotrophic lateral sclerosis patient iPSC-derived astrocytes impair autophagy via non-cell autonomous mechanisms. Mol. Brain 2017, 10, 1–12. [Google Scholar] [CrossRef]
  58. Ajmone-Cat, M.A.; Onori, A.; Toselli, C.; Stronati, E.; Morlando, M.; Bozzoni, I.; Monni, E.; Kokaia, Z.; Lupo, G.; Minghetti, L.; et al. Increased FUS levels in astrocytes leads to astrocyte and microglia activation and neuronal death. Sci. Rep. 2019, 9, 4572. [Google Scholar] [CrossRef]
  59. Sabatelli, M.; Moncada, A.; Conte, A.; Lattante, S.; Marangi, G.; Luigetti, M.; Lucchini, M.; Mirabella, M.; Romano, A.; del Grande, A.; et al. Mutations in the 3′ untranslated region of FUS causing FUS overexpression are associated with amyotrophic lateral sclerosis. Hum. Mol. Genet. 2013, 22, 4748–4755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Modigliani, S.D.; Morlando, M.; Errichelli, L.; Sabatelli, M.; Bozzoni, I. An ALS-associated mutation in the FUS 3′-UTR disrupts a microRNA–FUS regulatory circuitry. Nat. Commun. 2014, 5, 4335. [Google Scholar] [CrossRef]
  61. Aebischer, J.; Bernard-Marissal, N.; Pettmann, B.; Raoul, C. Death receptors in the selective degeneration of motoneurons in amyotrophic lateral sclerosis. J. Neurodegener. Dis. 2013, 2013, 746845. [Google Scholar] [CrossRef]
  62. Correia, A.S.; Patel, P.; Dutta, K.; Julien, J.-P. Inflammation induces TDP-43 mislocalization and aggregation. PLoS ONE 2015, 10, e0140248. [Google Scholar] [CrossRef] [Green Version]
  63. Herman, A.M.; Khandelwal, P.J.; Rebeck, G.W.; Moussa, C.E.-H. Wild type TDP-43 induces neuro-inflammation and alters APP metabolism in lentiviral gene transfer models. Exp. Neurol. 2012, 235, 297–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Zhao, W.; Beers, D.R.; Bell, S.; Wang, J.; Wen, S.; Baloh, R.H.; Appel, S.H. TDP-43 activates microglia through NF-κB and NLRP3 inflammasome. Exp. Neurol. 2015, 273, 24–35. [Google Scholar] [CrossRef]
  65. Smethurst, P.; Risse, E.; Tyzack, G.E.; Mitchell, J.S.; Taha, D.M.; Chen, Y.-R.; Newcombe, J.; Collinge, J.; Sidle, K.; Patani, R. Distinct responses of neurons and astrocytes to TDP-43 proteinopathy in amyotrophic lateral sclerosis. Brain 2020, 143, 430–440. [Google Scholar] [CrossRef]
  66. Fomin, V.; Richard, P.; Hoque, M.; Li, C.; Gu, Z.; Fissore-O’Leary, M.; Tian, B.; Prives, C.; Manley, J.L. The C9ORF72 gene, implicated in amyotrophic lateral sclerosis and frontotemporal dementia, encodes a protein that functions in control of endothelin and glutamate signaling. Mol. Cell. Biol. 2018, 38, e00155-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Rothstein, J.D.; Martin, L.; Levey, A.I.; Dykes-Hoberg, M.; Jin, L.; Wu, D.; Nash, N.; Kuncl, R.W. Localization of neuronal and glial glutamate transporters. Neuron 1994, 13, 713–725. [Google Scholar] [CrossRef]
  68. Rothstein, J.D.; Dykes-Hoberg, M.; Pardo, C.A.; Bristol, L.A.; Jin, L.; Kuncl, R.W.; Kanai, Y.; Hediger, M.A.; Wang, Y.; Schielke, J.P.; et al. Knockout of glutamate transporters reveals a major role for astroglial transport in excitotoxicity and clearance of glutamate. Neuron 1996, 16, 675–686. [Google Scholar] [CrossRef] [Green Version]
  69. Diaper, D.; Adachi, Y.; Lazarou, L.; Greenstein, M.; Simoes, F.; di Domenico, A.; Solomon, D.A.; Lowe, S.; Alsubaie, R.; Cheng, D.; et al. Drosophila TDP-43 dysfunction in glia and muscle cells cause cytological and behavioural phenotypes that characterize ALS and FTLD. Hum. Mol. Genet. 2013, 22, 3883–3893. [Google Scholar] [CrossRef] [Green Version]
  70. Estes, P.S.; Daniel, S.; Mccallum, A.P.; Boehringer, A.V.; Sukhina, A.S.; Zwick, R.A.; Zarnescu, D.C. Motor neurons and glia exhibit specific individualized responses to TDP-43 expression in a drosophila model of amyotrophic lateral sclerosis. Dis. Model. Mech. 2013, 6, 721–733. [Google Scholar] [CrossRef] [Green Version]
  71. Cragnaz, L.; Klima, R.; de Conti, L.; Romano, G.; Feiguin, F.; Buratti, E.; Baralle, M.; Baralle, F. An age-related reduction of brain TBPH/TDP-43 levels precedes the onset of locomotion defects in a drosophila ALS model. Neuroscience 2015, 311, 415–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Peng, A.Y.T.; Agrawal, I.; Ho, W.Y.; Yen, Y.-C.; Pinter, A.J.; Liu, J.; Phua, Q.X.C.; Koh, K.B.; Chang, J.-C.; Sanford, E.; et al. Loss of TDP-43 in astrocytes leads to motor deficits by triggering A1-like reactive phenotype and triglial dysfunction. Proc. Natl. Acad. Sci. USA 2020, 117, 29101–29112. [Google Scholar] [CrossRef] [PubMed]
  73. Kwiatkowski, T.J., Jr.; Bosco, D.A.; Leclerc, A.L.; Tamrazian, E.; Vanderburg, C.R.; Russ, C.; Davis, A.; Gilchrist, J.; Kasarskis, E.J.; Munsat, T.; et al. Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science 2009, 323, 1205–1208. [Google Scholar] [CrossRef] [Green Version]
  74. Sreedharan, J.; Blair, I.P.; Tripathi, V.B.; Hu, X.; Vance, C.; Rogelj, B.; Ackerley, S.; Durnall, J.C.; Williams, K.L.; Buratti, E.; et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science 2008, 319, 1668–1672. [Google Scholar] [CrossRef] [PubMed]
  75. Vance, C.; Rogelj, B.; Hortobágyi, T.; de Vos, K.J.; Nishimura, A.L.; Sreedharan, J.; Hu, X.; Smith, B.; Ruddy, D.; Wright, P.; et al. Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6. Science 2009, 323, 1208–1211. [Google Scholar] [CrossRef] [Green Version]
  76. Birsa, N.; Bentham, M.P.; Fratta, P. Cytoplasmic functions of TDP-43 and FUS and their role in ALS. Semin. Cell Dev. Biol. 2020, 99, 193–201. [Google Scholar] [CrossRef] [PubMed]
  77. Ederle, H.; Dormann, D. TDP-43 and FUS en route from the nucleus to the cytoplasm. FEBS Lett. 2017, 591, 1489–1507. [Google Scholar] [CrossRef] [Green Version]
  78. Nishihira, Y.; Tan, C.-F.; Onodera, O.; Toyoshima, Y.; Yamada, M.; Morita, T.; Nishizawa, M.; Kakita, A.; Takahashi, H. Sporadic amyotrophic lateral sclerosis: Two pathological patterns shown by analysis of distribution of TDP-43-immunoreactive neuronal and glial cytoplasmic inclusions. Acta Neuropathol. 2008, 116, 169–182. [Google Scholar] [CrossRef] [PubMed]
  79. Zhang, H.; Tan, C.-F.; Mori, F.; Tanji, K.; Kakita, A.; Takahashi, H.; Wakabayashi, K. TDP-43-immunoreactive neuronal and glial inclusions in the neostriatum in amyotrophic lateral sclerosis with and without dementia. Acta Neuropathol. 2007, 115, 115–122. [Google Scholar] [CrossRef]
  80. Arai, T.; Hasegawa, M.; Akiyama, H.; Ikeda, K.; Nonaka, T.; Mori, H.; Mann, D.; Tsuchiya, K.; Yoshida, M.; Hashizume, Y.; et al. TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem. Biophys. Res. Commun. 2006, 351, 602–611. [Google Scholar] [CrossRef] [PubMed]
  81. Neumann, M.; Sampathu, D.M.; Kwong, L.K.; Truax, A.C.; Micsenyi, M.C.; Chou, T.T.; Bruce, J.; Schuck, T.; Grossman, M.; Clark, C.M.; et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 2006, 314, 130–133. [Google Scholar] [CrossRef] [Green Version]
  82. Romano, G.; Appocher, C.; Scorzeto, M.; Klima, R.; Baralle, F.E.; Megighian, A.; Feiguin, F. Glial TDP-43 regulates axon wrapping, GluRIIA clustering and fly motility by autonomous and non-autonomous mechanisms. Hum. Mol. Genet. 2015, 24, 6134–6145. [Google Scholar] [CrossRef] [Green Version]
  83. Khalfallah, Y.; Kuta, R.; Grasmuck, C.; Prat, A.; Durham, H.D.; Velde, C.V. TDP-43 regulation of stress granule dynamics in neurodegenerative disease-relevant cell types. Sci. Rep. 2018, 8, 7551. [Google Scholar] [CrossRef] [Green Version]
  84. Moujalled, D.; Grubman, A.; Acevedo, K.; Yang, S.; Ke, Y.; Moujalled, D.M.; Duncan, C.; Caragounis, A.; Perera, N.D.; Turner, B.; et al. TDP-43 mutations causing amyotrophic lateral sclerosis are associated with altered expression of RNA-binding protein hnRNP K and affect the Nrf2 antioxidant pathway. Hum. Mol. Genet. 2017, 26, 1732–1746. [Google Scholar] [CrossRef]
  85. Barton, S.K.; Lau, C.L.; Chiam, M.D.F.; Tomas, D.; Muyderman, H.; Beart, P.M.; Turner, B.J. Mutant TDP-43 expression triggers TDP-43 pathology and cell autonomous effects on primary astrocytes: Implications for non-cell autonomous pathology in ALS. Neurochem. Res. 2020, 45, 1451–1459. [Google Scholar] [CrossRef]
  86. Velebit, J.; Horvat, A.; Smolič, T.; Mihevc, S.P.; Rogelj, B.; Zorec, R.; Vardjan, N. Astrocytes with TDP-43 inclusions exhibit reduced noradrenergic cAMP and Ca 2+ signaling and dysregulated cell metabolism. Sci. Rep. 2020, 10, 1–18. [Google Scholar] [CrossRef] [Green Version]
  87. Swarup, V.; Phaneuf, D.; Dupré, N.; Petri, S.; Strong, M.; Kriz, J.; Julien, J.-P. Deregulation of TDP-43 in amyotrophic lateral sclerosis triggers nuclear factor κB–mediated pathogenic pathways. J. Exp. Med. 2011, 208, 2429–2447. [Google Scholar] [CrossRef] [PubMed]
  88. Xia, Q.; Hu, Q.; Wang, H.; Yang, H.; Gao, F.; Ren, H.; Chen, D.; Fu, C.; Zheng, L.; Zhen, X.; et al. Induction of COX-2-PGE2 synthesis by activation of the MAPK/ERK pathway contributes to neuronal death triggered by TDP-43-depleted microglia. Cell Death Dis. 2015, 6, e1702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. LaRocca, T.J.; Mariani, A.; Watkins, L.R.; Link, C.D. TDP-43 knockdown causes innate immune activation via protein kinase R in astrocytes. Neurobiol. Dis. 2019, 132, 104514. [Google Scholar] [CrossRef] [PubMed]
  90. Kasai, T.; Tokuda, T.; Ishigami, N.; Sasayama, H.; Foulds, P.; Mitchell, D.J.; Mann, D.M.A.; Allsop, D.; Nakagawa, M. Increased TDP-43 protein in cerebrospinal fluid of patients with amyotrophic lateral sclerosis. Acta Neuropathol. 2009, 117, 55–62. [Google Scholar] [CrossRef]
  91. Noto, Y.-I.; Shibuya, K.; Sato, Y.; Kanai, K.; Misawa, S.; Sawai, S.; Mori, M.; Uchiyama, T.; Isose, S.; Nasu, S.; et al. Elevated CSF TDP-43 levels in amyotrophic lateral sclerosis: Specificity, sensitivity, and a possible prognostic value. Amyotroph. Lateral Scler. 2010, 12, 140–143. [Google Scholar] [CrossRef] [PubMed]
  92. Steinacker, P.; Hendrich, C.; Sperfeld, A.D.; Jesse, S.; von Arnim, C.A.F.; Lehnert, S.; Pabst, A.; Uttner, I.; Tumani, H.; Lee, V.M.-Y.; et al. TDP-43 in cerebrospinal fluid of patients with frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Arch. Neurol. 2008, 65, 1481–1487. [Google Scholar] [CrossRef] [Green Version]
  93. Smethurst, P.; Sidle, K.C.L.; Hardy, J. Review: Prion-like mechanisms of transactive response DNA binding protein of 43 kDa (TDP-43) in amyotrophic lateral sclerosis (ALS). Neuropathol. Appl. Neurobiol. 2014, 41, 578–597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Leal-Lasarte, M.M.; Franco, J.M.; Labrador-Garrido, A.; Pozo, D.; Roodveldt, C. Extracellular TDP-43 aggregates target MAPK/MAK/MRK overlapping kinase (MOK) and trigger caspase-3/IL-18 signaling in microglia. FASEB J. 2017, 31, 2797–2816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Smeyers, J.; Banchi, E.-G.; Latouche, M. C9ORF72: What it is, what it does, and why it matters. Front. Cell. Neurosci. 2021, 15, 661447. [Google Scholar] [CrossRef]
  96. Farg, M.A.; Sundaramoorthy, V.; Sultana, J.M.; Yang, S.; Atkinson, R.; Levina, V.; Halloran, M.A.; Gleeson, P.; Blair, I.; Soo, K.Y.; et al. C9ORF72, implicated in amytrophic lateral sclerosis and frontotemporal dementia, regulates endosomal trafficking. Hum. Mol. Genet. 2014, 23, 3579–3595. [Google Scholar] [CrossRef]
  97. Levine, T.P.; Daniels, R.D.; Gatta, A.T.; Wong, L.H.; Hayes, M.J. The product of C9orf72, a gene strongly implicated in neurodegeneration, is structurally related to DENN Rab-GEFs. Bioinformatics 2013, 29, 499–503. [Google Scholar] [CrossRef] [Green Version]
  98. Sellier, C.; Campanari, M.; Corbier, C.J.; Gaucherot, A.; Kolb-Cheynel, I.; Oulad-Abdelghani, M.; Ruffenach, F.; Page, A.; Ciura, S.; Kabashi, E.; et al. Loss of C9 ORF 72 impairs autophagy and synergizes with polyQ Ataxin-2 to induce motor neuron dysfunction and cell death. EMBO J. 2016, 35, 1276–1297. [Google Scholar] [CrossRef] [PubMed]
  99. Sullivan, P.M.; Zhou, X.; Robins, A.M.; Paushter, D.H.; Kim, D.; Smolka, M.B.; Hu, F. The ALS/FTLD associated protein C9orf72 associates with SMCR8 and WDR41 to regulate the autophagy-lysosome pathway. Acta Neuropathol. Commun. 2016, 4, 51. [Google Scholar] [CrossRef] [Green Version]
  100. Webster, C.P.; Smith, E.F.; Bauer, C.S.; Moller, A.; Hautbergue, G.M.; Ferraiuolo, L.; Myszczynska, M.; Higginbottom, A.; Walsh, M.J.; Whitworth, A.J.; et al. The C9orf72 protein interacts with Rab1a and the ULK 1 complex to regulate initiation of autophagy. EMBO J. 2016, 35, 1656–1676. [Google Scholar] [CrossRef]
  101. Webster, C.P.; Smith, E.F.; Grierson, A.J.; de Vos, K.J. C9orf72 plays a central role in Rab GTPase-dependent regulation of autophagy. Small GTPases 2018, 9, 399–408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Yang, M.; Liang, C.; Swaminathan, K.; Herrlinger, S.; Lai, F.; Shiekhattar, R.; Chen, J.-F. A C9ORF72/SMCR8-containing complex regulates ULK1 and plays a dual role in autophagy. Sci. Adv. 2016, 2, e1601167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Zhang, D.; Iyer, L.M.; He, F.; Aravind, L. Discovery of novel DENN proteins: Implications for the evolution of eukaryotic intracellular membrane structures and human disease. Front. Genet. 2012, 3, 283. [Google Scholar] [CrossRef] [Green Version]
  104. Atanasio, A.; Decman, V.; White, D.; Ramos, M.; Ikiz, B.; Lee, H.-C.; Siao, C.-J.; Brydges, S.; LaRosa, E.; Bai, Y.; et al. C9orf72 ablation causes immune dysregulation characterized by leukocyte expansion, autoantibody production and glomerulonephropathy in mice. Sci. Rep. 2016, 6, 23204. [Google Scholar] [CrossRef] [PubMed]
  105. Burberry, A.; Suzuki, N.; Wang, J.-Y.; Moccia, R.; Mordes, D.A.; Stewart, M.H.; Suzuki-Uematsu, S.; Ghosh, S.; Singh, A.; Merkle, F.T.; et al. Loss-of-function mutations in the C9ORF72 mouse ortholog cause fatal autoimmune disease. Sci. Transl. Med. 2016, 8, 347ra93. [Google Scholar] [CrossRef] [Green Version]
  106. O’Rourke, J.G.; Bogdanik, L.; Yáñez, A.; Lall, D.; Wolf, A.J.; Muhammad, A.K.M.G.; Ho, R.; Carmona, S.; Vit, J.P.; Zarrow, J.; et al. C9orf72 is required for proper macrophage and microglial function in mice. Science 2016, 351, 1324–1329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. O’Rourke, J.G.; Bogdanik, L.; Muhammad, A.; Gendron, T.F.; Kim, K.J.; Austin, A.; Cady, J.; Liu, E.Y.; Zarrow, J.; Grant, S.; et al. C9orf72 BAC transgenic mice display typical pathologic features of ALS/FTD. Neuron 2015, 88, 892–901. [Google Scholar] [CrossRef] [Green Version]
  108. Peters, O.; Cabrera, G.T.; Tran, H.; Gendron, T.F.; McKeon, J.E.; Metterville, J.; Weiss, A.; Wightman, N.; Salameh, J.; Kim, J.; et al. Human C9ORF72 hexanucleotide expansion reproduces RNA foci and dipeptide repeat proteins but not neurodegeneration in BAC transgenic mice. Neuron 2015, 88, 902–909. [Google Scholar] [CrossRef] [Green Version]
  109. Shi, Y.; Lin, S.; Staats, K.A.; Li, Y.; Chang, W.-H.; Hung, S.-T.; Hendricks, E.; Linares, G.R.; Wang, Y.; Son, E.Y.; et al. Haploinsufficiency leads to neurodegeneration in C9ORF72 ALS/FTD human induced motor neurons. Nat. Med. 2018, 24, 313–325. [Google Scholar] [CrossRef]
  110. Langseth, A.; Kim, J.; Ugolino, J.E.; Shah, Y.; Hwang, H.-Y.; Wang, J.; Bergles, D.E.; Brown, S.P. Cell-type specific differences in promoter activity of the ALS-linked C9orf72 mouse ortholog. Sci. Rep. 2017, 7, 1–13. [Google Scholar] [CrossRef]
  111. Braems, E.; Swinnen, B.; Bosch, L.V.D. C9orf72 loss-of-function: A trivial, stand-alone or additive mechanism in C9 ALS/FTD? Acta Neuropathol. 2020, 140, 625–643. [Google Scholar] [CrossRef] [PubMed]
  112. Al-Sarraj, S.; King, A.; Troakes, C.; Smith, B.; Maekawa, S.; Bodi, I.; Rogelj, B.; Al-Chalabi, A.; Hortobágyi, T.; Shaw, C.E. P62 positive, TDP-43 negative, neuronal cytoplasmic and intranuclear inclusions in the cerebellum and hippocampus define the pathology of C9orf72-linked FTLD and MND/ALS. Acta Neuropathol. 2011, 122, 691–702. [Google Scholar] [CrossRef]
  113. Balendra, R.; Isaacs, A.M. C9orf72-mediated ALS and FTD: Multiple pathways to disease. Nat. Rev. Neurol. 2018, 14, 544–558. [Google Scholar] [CrossRef]
  114. Rostalski, H.; Leskelä, S.; Huber, N.; Katisko, K.; Cajanus, A.; Solje, E.; Marttinen, M.; Natunen, T.; Remes, A.M.; Hiltunen, M.; et al. Astrocytes and microglia as potential contributors to the pathogenesis of C9orf72 repeat expansion-associated FTLD and ALS. Front. Neurosci. 2019, 13, 486. [Google Scholar] [CrossRef]
  115. Cooper-Knock, J.; Walsh, M.J.; Higginbottom, A.; Highley, R.; Dickman, M.; Edbauer, D.; Ince, P.G.; Wharton, S.B.; Wilson, S.; Kirby, J.; et al. Sequestration of multiple RNA recognition motif-containing proteins by C9orf72 repeat expansions. Brain 2014, 137, 2040–2051. [Google Scholar] [CrossRef]
  116. Donnelly, C.J.; Zhang, P.-W.; Pham, J.T.; Haeusler, A.R.; Mistry, N.A.; Vidensky, S.; Daley, E.; Poth, E.M.; Hoover, B.; Fines, D.M.; et al. RNA toxicity from the ALS/FTD C9ORF72 expansion is mitigated by antisense intervention. Neuron 2013, 80, 415–428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Hautbergue, G.M.; Castelli, L.M.; Ferraiuolo, L.; Sanchez-Martinez, A.; Cooper-Knock, J.; Higginbottom, A.; Lin, Y.-H.; Bauer, C.S.; Dodd, J.E.; Myszczynska, M.; et al. SRSF1-dependent nuclear export inhibition of C9ORF72 repeat transcripts prevents neurodegeneration and associated motor deficits. Nat. Commun. 2017, 8, 16063. [Google Scholar] [CrossRef]
  118. Mori, K.; Lammich, S.; MacKenzie, I.R.A.; Forné, I.; Zilow, S.; Kretzschmar, H.; Edbauer, D.; Janssens, J.; Kleinberger, G.; Cruts, M.; et al. HnRNP A3 binds to GGGGCC repeats and is a constituent of p62-positive/TDP43-negative inclusions in the hippocampus of patients with C9orf72 mutations. Acta Neuropathol. 2013, 125, 413–423. [Google Scholar] [CrossRef]
  119. Ramesh, N.; Daley, E.L.; Gleixner, A.M.; Mann, J.R.; Kour, S.; Mawrie, D.; Anderson, E.N.; Kofler, J.; Donnelly, C.J.; Kiskinis, E.; et al. RNA dependent suppression of C9orf72 ALS/FTD associated neurodegeneration by matrin-3. Acta Neuropathol. Commun. 2020, 8, 1–20. [Google Scholar] [CrossRef]
  120. Rossi, S.; Serrano, A.; Gerbino, V.; Giorgi, A.; di Francesco, L.; Nencini, M.; Bozzo, F.; Schininà, M.E.; Bagni, C.; Cestra, G.; et al. Nuclear accumulation of mRNAs underlies G4C2 repeat-induced translational repression in a cellular model of C9orf72 ALS. J. Cell Sci. 2015, 128, 1787–1799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Sareen, D.; O’Rourke, J.G.; Meera, P.; Muhammad, A.K.M.G.; Grant, S.; Simpkinson, M.; Bell, S.; Carmona, S.; Ornelas, L.; Sahabian, A.; et al. Targeting RNA foci in iPSC-derived motor neurons from ALS patients with a C9ORF72 repeat expansion. Sci. Transl. Med. 2013, 5, 208ra149. [Google Scholar] [CrossRef] [Green Version]
  122. Conlon, E.G.; Lu, L.; Sharma, A.; Yamazaki, T.; Tang, T.; Shneider, N.A.; Manley, J.L. The C9ORF72 GGGGCC expansion forms RNA G-quadruplex inclusions and sequesters hnRNP H to disrupt splicing in ALS brains. eLife 2016, 5, e17820. [Google Scholar] [CrossRef] [PubMed]
  123. Lee, Y.-B.; Chen, H.-J.; Peres, J.N.; Gomez-Deza, J.; Attig, J.; Štalekar, M.; Troakes, C.; Nishimura, A.L.; Scotter, E.L.; Vance, C.; et al. Hexanucleotide repeats in ALS/FTD form length-dependent RNA foci, sequester RNA binding proteins, and are neurotoxic. Cell Rep. 2013, 5, 1178–1186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Conlon, E.G.; Fagegaltier, D.; Agius, P.; Davis-Porada, J.; Gregory, J.; Hubbard, I.; Kang, K.; Kim, D.; Phatnani, H.; Kwan, J.; et al. Unexpected similarities between C9ORF72 and sporadic forms of ALS/FTD suggest a common disease mechanism. eLife 2018, 7, e37754. [Google Scholar] [CrossRef]
  125. Prudencio, M.; Belzil, V.V.; Batra, R.; Ross, C.A.; Gendron, T.F.; Pregent, L.J.; Murray, M.E.; Overstreet, K.K.; Piazza-Johnston, A.E.; Desaro, P.; et al. Distinct brain transcriptome profiles in C9orf72-associated and sporadic ALS. Nat. Neurosci. 2015, 18, 1175–1182. [Google Scholar] [CrossRef]
  126. Goodman, L.; Bonini, N.M. Repeat-associated non-AUG (RAN) translation mechanisms are running into focus for GGGGCC-repeat associated ALS/FTD. Prog. Neurobiol. 2019, 183, 101697. [Google Scholar] [CrossRef]
  127. Schmitz, A.; Marques, J.P.; Oertig, I.; Maharjan, N.; Saxena, S. Emerging perspectives on dipeptide repeat proteins in C9ORF72 ALS/FTD. Front. Cell. Neurosci. 2021, 15, 637548. [Google Scholar] [CrossRef]
  128. Saberi, S.; Stauffer, J.E.; Jiang, J.; Garcia, S.D.; Taylor, A.E.; Schulte, D.; Ohkubo, T.; Schloffman, C.L.; Maldonado, M.; Baughn, M.; et al. Sense-encoded poly-GR dipeptide repeat proteins correlate to neurodegeneration and uniquely co-localize with TDP-43 in dendrites of repeat-expanded C9orf72 amyotrophic lateral sclerosis. Acta Neuropathol. 2018, 135, 459–474. [Google Scholar] [CrossRef]
  129. Schipper, L.J.; Raaphorst, J.; Aronica, E.; Baas, F.; de Haan, R.J.; de Visser, M.; Troost, D. Prevalence of brain and spinal cord inclusions, including dipeptide repeat proteins, in patients with the C9ORF72 hexanucleotide repeat expansion: A systematic neuropathological review. Neuropathol. Appl. Neurobiol. 2016, 42, 547–560. [Google Scholar] [CrossRef] [Green Version]
  130. Root, J.; Merino, P.; Nuckols, A.; Johnson, M.; Kukar, T. Lysosome dysfunction as a cause of neurodegenerative diseases: Lessons from frontotemporal dementia and amyotrophic lateral sclerosis. Neurobiol. Dis. 2021, 154, 105360. [Google Scholar] [CrossRef] [PubMed]
  131. Allen, S.; Hall, B.; Castelli, L.M.; Francis, L.; Woof, R.; Siskos, A.; Kouloura, E.; Gray, E.; Thompson, A.G.; Talbot, K.; et al. Astrocyte adenosine deaminase loss increases motor neuron toxicity in amyotrophic lateral sclerosis. Brain 2019, 142, 586–605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Allen, S.P.; Hall, B.; Woof, R.; Francis, L.; Gatto, N.; Shaw, A.C.; Myszczynska, M.; Hemingway, J.; Coldicott, I.; Willcock, A.; et al. C9orf72 expansion within astrocytes reduces metabolic flexibility in amyotrophic lateral sclerosis. Brain 2019, 142, 3771–3790. [Google Scholar] [CrossRef]
  133. Zaepfel, B.L.; Rothstein, J.D. RNA is a double-edged sword in ALS pathogenesis. Front. Cell. Neurosci. 2021, 15, 708181. [Google Scholar] [CrossRef]
  134. Arnold, E.S.; Ling, S.-C.; Huelga, S.C.; Lagier-Tourenne, C.; Polymenidou, M.; Ditsworth, D.; Kordasiewicz, H.B.; McAlonis-Downes, M.; Platoshyn, O.; Parone, P.A.; et al. ALS-linked TDP-43 mutations produce aberrant RNA splicing and adult-onset motor neuron disease without aggregation or loss of nuclear TDP-43. Proc. Natl. Acad. Sci. USA 2013, 110, E736–E745. [Google Scholar] [CrossRef] [Green Version]
  135. Donde, A.; Sun, M.; Ling, J.; Braunstein, K.E.; Pang, B.; Wen, X.; Cheng, X.; Chen, L.; Wong, P.C. Splicing repression is a major function of TDP-43 in motor neurons. Acta Neuropathol. 2019, 138, 813–826. [Google Scholar] [CrossRef]
  136. Fratta, P.; Sivakumar, P.; Humphrey, J.; Lo, K.; Ricketts, T.; Oliveira, H.; Brito-Armas, J.M.; Kalmar, B.; Ule, A.; Yu, Y.; et al. Mice with endogenous TDP -43 mutations exhibit gain of splicing function and characteristics of amyotrophic lateral sclerosis. EMBO J. 2018, 37. [Google Scholar] [CrossRef]
  137. Jutzi, D.; Campagne, S.; Schmidt, R.; Reber, S.; Mechtersheimer, J.; Gypas, F.; Schweingruber, C.; Colombo, M.; von Schroetter, C.; Loughlin, F.E.; et al. Aberrant interaction of FUS with the U1 snRNA provides a molecular mechanism of FUS induced amyotrophic lateral sclerosis. Nat. Commun. 2020, 11, 1–14. [Google Scholar] [CrossRef] [PubMed]
  138. Ling, J.P.; Pletnikova, O.; Troncoso, J.C.; Wong, P.C. TDP-43 repression of nonconserved cryptic exons is compromised in ALS-FTD. Science 2015, 349, 650–655. [Google Scholar] [CrossRef] [Green Version]
  139. Reber, S.; Stettler, J.; Filosa, G.; Colombo, M.; Jutzi, D.; Lenzken, S.C.; Schweingruber, C.; Bruggmann, R.; Bachi, A.; Barabino, S.M.; et al. Minor intron splicing is regulated by FUS and affected by ALS -associated FUS mutants. EMBO J. 2016, 35, 1504–1521. [Google Scholar] [CrossRef]
  140. Sun, S.; Ling, S.-C.; Qiu, J.; Albuquerque, C.P.; Zhou, Y.; Tokunaga, S.; Li, H.; Qiu, H.; Bui, A.; Yeo, E.; et al. ALS-causative mutations in FUS/TLS confer gain and loss of function by altered association with SMN and U1-snRNP. Nat. Commun. 2015, 6, 1–14. [Google Scholar] [CrossRef] [PubMed]
  141. Wang, Q.; Conlon, E.G.; Manley, J.L.; Rio, D.C. Widespread intron retention impairs protein homeostasis in C9orf72 ALS brains. Genome Res. 2020, 30, 1705–1715. [Google Scholar] [CrossRef] [PubMed]
  142. White, M.A.; Kim, E.; Duffy, A.; Adalbert, R.; Phillips, B.U.; Peters, O.M.; Stephenson, J.; Yang, S.; Massenzio, F.; Lin, Z.; et al. TDP-43 gains function due to perturbed autoregulation in a Tardbp knock-in mouse model of ALS-FTD. Nat. Neurosci. 2018, 21, 552–563. [Google Scholar] [CrossRef] [PubMed]
  143. Yin, S.; Lopez-Gonzalez, R.; Kunz, R.C.; Gangopadhyay, J.; Borufka, C.; Gygi, S.P.; Gao, F.-B.; Reed, R. Evidence that C9ORF72 dipeptide repeat proteins associate with U2 snRNP to cause mis-splicing in ALS/FTD Patients. Cell Rep. 2017, 19, 2244–2256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Kwon, I.; Xiang, S.; Kato, M.; Wu, L.; Theodoropoulos, P.; Wang, T.; Kim, J.; Yun, J.; Xie, Y.; McKnight, S.L. Poly-dipeptides encoded by the C9orf72 repeats bind nucleoli, impede RNA biogenesis, and kill cells. Science 2014, 345, 1139–1145. [Google Scholar] [CrossRef] [Green Version]
  145. Lin, C.-L.G.; Bristol, L.A.; Jin, L.; Dykes-Hoberg, M.; Crawford, T.; Clawson, L.; Rothstein, J.D. Aberrant RNA processing in a neurodegenerative disease: The cause for absent EAAT2, a glutamate transporter, in amyotrophic lateral sclerosis. Neuron 1998, 20, 589–602. [Google Scholar] [CrossRef] [Green Version]
  146. Humphrey, J.; Birsa, N.; Milioto, C.; McLaughlin, M.; Ule, A.M.; Robaldo, D.; Eberle, A.B.; Kräuchi, R.; Bentham, M.; Brown, A.-L.; et al. FUS ALS-causative mutations impair FUS autoregulation and splicing factor networks through intron retention. Nucleic Acids Res. 2020, 48, 6889–6905. [Google Scholar] [CrossRef]
  147. Fratta, P.; Isaacs, A.M. The snowball effect of RNA binding protein dysfunction in amyotrophic lateral sclerosis. Brain 2018, 141, 1236–1238. [Google Scholar] [CrossRef] [Green Version]
  148. Johnson, J.O.; Mandrioli, J.; Benatar, M.; Abramzon, Y.; van Deerlin, V.M.; Trojanowski, J.Q.; Gibbs, J.R.; Brunetti, M.; Gronka, S.; Wuu, J.; et al. Exome sequencing reveals VCP mutations as a cause of familial ALS. Neuron 2010, 68, 857–864. [Google Scholar] [CrossRef] [Green Version]
  149. Ziff, O.J.; Taha, D.M.; Crerar, H.; Clarke, B.E.; Chakrabarti, A.M.; Kelly, G.; Neeves, J.; Tyzack, G.E.; Luscombe, N.M.; Patani, R. Reactive astrocytes in ALS display diminished intron retention. Nucleic Acids Res. 2021, 49, 3168–3184. [Google Scholar] [CrossRef] [PubMed]
  150. Luisier, R.; Tyzack, G.E.; Hall, C.E.; Mitchell, J.S.; Devine, H.; Taha, D.M.; Malik, B.; Meyer, I.; Greensmith, L.; Newcombe, J.; et al. Intron retention and nuclear loss of SFPQ are molecular hallmarks of ALS. Nat. Commun. 2018, 9, 1–15. [Google Scholar] [CrossRef]
  151. Tyzack, G.E.; Neeves, J.; Crerar, H.; Klein, P.; Ziff, O.; Taha, D.M.; Luisier, R.; Luscombe, N.M.; Patani, R. Aberrant cytoplasmic intron retention is a blueprint for RNA binding protein mislocalization in VCP-related amyotrophic lateral sclerosis. Brain 2021, 144, 1985–1993. [Google Scholar] [CrossRef]
  152. Lee, J.; Villarreal, O.D.; Chen, X.; Zandee, S.; Young, Y.K.; Torok, C.; Lamarche-Vane, N.; Prat, A.; Rivest, S.; Gosselin, D.; et al. QUAKING regulates microexon alternative splicing of the rho GTPase pathway and controls microglia homeostasis. Cell Rep. 2020, 33, 108560. [Google Scholar] [CrossRef]
  153. Cestra, G.; Rossi, S.; di Salvio, M.; Cozzolino, M. Control of mRNA translation in ALS proteinopathy. Front. Mol. Neurosci. 2017, 10, 85. [Google Scholar] [CrossRef] [PubMed]
  154. Lehmkuhl, E.M.; Zarnescu, D.C. Lost in translation: Evidence for protein synthesis deficits in ALS/FTD and related neurodegenerative diseases. Adv. Neurobiol. 2018, 20, 283–301. [Google Scholar] [CrossRef] [PubMed]
  155. Frottin, F.; Pérez-Berlanga, M.; Hartl, F.U.; Hipp, M.S. Multiple pathways of toxicity induced by C9orf72 dipeptide repeat aggregates and G4C2 RNA in a cellular model. eLife 2021, 10, e62718. [Google Scholar] [CrossRef]
  156. Hartmann, H.; Hornburg, D.; Czuppa, M.; Bader, J.; Michaelsen, M.; Farny, D.; Arzberger, T.; Mann, M.; Meissner, F.; Edbauer, D. Proteomics and C9orf72 neuropathology identify ribosomes as poly-GR/PR interactors driving toxicity. Life Sci. Alliance 2018, 1, e201800070. [Google Scholar] [CrossRef] [Green Version]
  157. Moens, T.G.; Niccoli, T.; Wilson, K.M.; Atilano, M.; Birsa, N.; Gittings, L.; Holbling, B.V.; Dyson, M.; Thoeng, A.; Neeves, J.; et al. C9orf72 arginine-rich dipeptide proteins interact with ribosomal proteins in vivo to induce a toxic translational arrest that is rescued by eIF1A. Acta Neuropathol. 2019, 137, 487–500. [Google Scholar] [CrossRef] [Green Version]
  158. Sun, Y.; Eshov, A.; Zhou, J.; Isiktas, A.U.; Guo, J.U. C9orf72 arginine-rich dipeptide repeats inhibit UPF1-mediated RNA decay via translational repression. Nat. Commun. 2020, 11, 3354. [Google Scholar] [CrossRef] [PubMed]
  159. Kanekura, K.; Yagi, T.; Cammack, A.J.; Mahadevan, J.; Kuroda, M.; Harms, M.B.; Miller, T.M.; Urano, F. Poly-dipeptides encoded by the C9ORF72 repeats block global protein translation. Hum. Mol. Genet. 2016, 25, 1803–1813. [Google Scholar] [CrossRef] [Green Version]
  160. López-Erauskin, J.; Tadokoro, T.; Baughn, M.W.; Myers, B.; McAlonis-Downes, M.; Chillon-Marinas, C.; Asiaban, J.N.; Artates, J.; Bui, A.T.; Vetto, A.P.; et al. ALS/FTD-linked mutation in FUS suppresses intra-axonal protein synthesis and drives disease without nuclear loss-of-function of FUS. Neuron 2020, 106, 354. [Google Scholar] [CrossRef] [PubMed]
  161. Birsa, N.; Ule, A.M.; Garone, M.G.; Tsang, B.; Mattedi, F.; Chong, P.A.; Humphrey, J.; Jarvis, S.; Pisiren, M.; Wilkins, O.G.; et al. FUS-ALS mutants alter FMRP phase separation equilibrium and impair protein translation. Sci. Adv. 2021, 7. [Google Scholar] [CrossRef]
  162. Garone, M.G.; Birsa, N.; Rosito, M.; Salaris, F.; Mochi, M.; de Turris, V.; Nair, R.R.; Cunningham, T.J.; Fisher, E.M.C.; Morlando, M.; et al. ALS-related FUS mutations alter axon growth in motoneurons and affect HuD/ELAVL4 and FMRP activity. Commun. Biol. 2021, 4, 1–17. [Google Scholar] [CrossRef] [PubMed]
  163. Lai, A.; Valdez-Sinon, A.N.; Bassell, G.J. Regulation of RNA granules by FMRP and implications for neurological diseases. Traffic 2020, 21, 454–462. [Google Scholar] [CrossRef]
  164. Higashimori, H.; Morel, L.; Huth, J.; Lindemann, L.; Dulla, C.; Taylor, A.; Freeman, M.; Yang, Y. Astroglial FMRP-dependent translational down-regulation of mGluR5 underlies glutamate transporter GLT1 dysregulation in the fragile X mouse. Hum. Mol. Genet. 2013, 22, 2041–2054. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Sakers, K.; Lake, A.M.; Khazanchi, R.; Ouwenga, R.; Vasek, M.J.; Dani, A.; Dougherty, J.D. Astrocytes locally translate transcripts in their peripheral processes. Proc. Natl. Acad. Sci. USA 2017, 114, E3830–E3838. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Non-cell-autonomous mechanisms underlying motor neuron degeneration mediated by ALS-linked C9orf72, FUS, or TDP-43 astrocytes and microglia. Wild-type motor neurons undergo cell death when co-cultured with astrocytes or microglia expressing ALS-linked C9orf72, FUS, or TDP-43. Mechanisms underlying non-cell-autonomous toxicity include loss of action potential output, altered expression levels of AMPA receptor subunits GluA1 and GluA2, and defects in neuromuscular junction (NMJ) morphology. These effects could be mediated by the extracellular secretion from glial cells of proinflammatory cytokines, such as IL-1β, IL-6, and TNFa, or TDP-43 aggregates.
Figure 1. Non-cell-autonomous mechanisms underlying motor neuron degeneration mediated by ALS-linked C9orf72, FUS, or TDP-43 astrocytes and microglia. Wild-type motor neurons undergo cell death when co-cultured with astrocytes or microglia expressing ALS-linked C9orf72, FUS, or TDP-43. Mechanisms underlying non-cell-autonomous toxicity include loss of action potential output, altered expression levels of AMPA receptor subunits GluA1 and GluA2, and defects in neuromuscular junction (NMJ) morphology. These effects could be mediated by the extracellular secretion from glial cells of proinflammatory cytokines, such as IL-1β, IL-6, and TNFa, or TDP-43 aggregates.
Cells 10 03005 g001
Figure 2. Features of RBP/RNA dysfunction in FUS and TDP-43 motor neurons, astrocytes, and microglia. FUS and TDP-43 are localized in the nucleus of normal cells, where they control several aspects of nuclear mRNA metabolism, including mRNA transcription and splicing. They also localize in the cytoplasm of cells where they participate in the control of mRNA transport and translation. In ALS motor neurons, FUS and TDP-43 massively delocalize in the cytoplasm, where they form cytoplasmic aggregates which frequently colocalize with stress granules (SGs). In these conditions, both gain of cytoplasmic toxic functions and loss of nuclear function contribute to the pathological mechanisms underlying motor neuron degeneration. In activated astrocytes and microglia, which are deeply involved in the progressive demise of motor neurons in ALS, cytoplasmic mislocalization of FUS and TDP-43 also occurs. In astrocytes, this leads to decreased intron retention in transcripts that regulate their reactive state, which eventually leads to an increase in their expression. Further, cytoplasmic ALS RBPs might interfere with the activity of FMRP causing alterations in local translation, which also occur in astrocyte distal processes. Dysregulation in the alternative splicing of proinflammatory transcripts also occurs in RBP-defective microglia thus supporting a role of these mechanisms in microglia activation in ALS.
Figure 2. Features of RBP/RNA dysfunction in FUS and TDP-43 motor neurons, astrocytes, and microglia. FUS and TDP-43 are localized in the nucleus of normal cells, where they control several aspects of nuclear mRNA metabolism, including mRNA transcription and splicing. They also localize in the cytoplasm of cells where they participate in the control of mRNA transport and translation. In ALS motor neurons, FUS and TDP-43 massively delocalize in the cytoplasm, where they form cytoplasmic aggregates which frequently colocalize with stress granules (SGs). In these conditions, both gain of cytoplasmic toxic functions and loss of nuclear function contribute to the pathological mechanisms underlying motor neuron degeneration. In activated astrocytes and microglia, which are deeply involved in the progressive demise of motor neurons in ALS, cytoplasmic mislocalization of FUS and TDP-43 also occurs. In astrocytes, this leads to decreased intron retention in transcripts that regulate their reactive state, which eventually leads to an increase in their expression. Further, cytoplasmic ALS RBPs might interfere with the activity of FMRP causing alterations in local translation, which also occur in astrocyte distal processes. Dysregulation in the alternative splicing of proinflammatory transcripts also occurs in RBP-defective microglia thus supporting a role of these mechanisms in microglia activation in ALS.
Cells 10 03005 g002
Figure 3. Contribution of loss- and gain-of-function mechanisms to the inflammatory processes mediated by C9orf72-ALS astrocytes and microglia. In ALS astrocytes and microglia, the loss of C9ORF72 protein induced by the presence of expanded hexanucleotide repeats (HRE) in the first intron of the C9orf72 gene, impacts the regulation of autophagy and endosome trafficking, and is linked to the upregulation of proinflammatory cytokines, altered immune responses, and lysosome accumulation which occur in ALS microglia and astrocytes. HRE-containing RNAs and dipeptide protein repeats (DPRs) that might accumulate in ALS glia also interfere with the regulation of splicing and translation by affecting the activities of RBPs involved in the control of RNA processing, including hnRNPH and TDP-43, and/or by interfering with ribosome biosynthesis and activity.
Figure 3. Contribution of loss- and gain-of-function mechanisms to the inflammatory processes mediated by C9orf72-ALS astrocytes and microglia. In ALS astrocytes and microglia, the loss of C9ORF72 protein induced by the presence of expanded hexanucleotide repeats (HRE) in the first intron of the C9orf72 gene, impacts the regulation of autophagy and endosome trafficking, and is linked to the upregulation of proinflammatory cytokines, altered immune responses, and lysosome accumulation which occur in ALS microglia and astrocytes. HRE-containing RNAs and dipeptide protein repeats (DPRs) that might accumulate in ALS glia also interfere with the regulation of splicing and translation by affecting the activities of RBPs involved in the control of RNA processing, including hnRNPH and TDP-43, and/or by interfering with ribosome biosynthesis and activity.
Cells 10 03005 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rossi, S.; Cozzolino, M. Dysfunction of RNA/RNA-Binding Proteins in ALS Astrocytes and Microglia. Cells 2021, 10, 3005. https://doi.org/10.3390/cells10113005

AMA Style

Rossi S, Cozzolino M. Dysfunction of RNA/RNA-Binding Proteins in ALS Astrocytes and Microglia. Cells. 2021; 10(11):3005. https://doi.org/10.3390/cells10113005

Chicago/Turabian Style

Rossi, Simona, and Mauro Cozzolino. 2021. "Dysfunction of RNA/RNA-Binding Proteins in ALS Astrocytes and Microglia" Cells 10, no. 11: 3005. https://doi.org/10.3390/cells10113005

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

Article Metrics

Back to TopTop