Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Dec 1.
Published in final edited form as: Neurobiol Dis. 2019 Aug 2;132:104562. doi: 10.1016/j.nbd.2019.104562

Examining the relationship between astrocyte dysfunction and neurodegeneration in ALS using hiPSCs

Madeline Halpern 1, Kristen J Brennand 2,3,4,5,#, James Gregory 6,#
PMCID: PMC6834907  NIHMSID: NIHMS1537563  PMID: 31381978

Abstract

Amyotrophic lateral sclerosis (ALS) is a complex and fatal neurodegenerative disease for which the causes of disease onset and progression remain unclear. Recent advances in human induced pluripotent stem cell (hiPSC)-based models permit the study of the genetic factors associated with ALS in patient-derived neural cell types, including motor neurons and glia. While astrocyte dysfunction has traditionally been thought to exacerbate disease progression, astrocytic dysfunction may play a more direct role in disease initiation and progression. Such non-cell autonomous mechanisms expand the potential targets of therapeutic intervention, but only a handful of ALS risk-associated genes have been examined for their impact on astrocyte dysfunction and neurodegeneration. This review summarizes what is currently known about astrocyte function in ALS and suggests ways in which hiPSC-based models can be used to more effectively study the role of astrocytes in neurodegenerative disease.

Keywords: Amyotrophic lateral sclerosis, astrocytes, human induced pluripotent stem cells

Introduction

The prevalence of age-related neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), continues to increase (Arthur et al., 2016; Heemels, 2016). Unfortunately, the primary mechanisms that drive the onset and progression of ALS are poorly understood, and current therapeutics provide minimal benefit. Here we present a case for increased focus on non-cell autonomous disease mechanisms arising from glial dysfunction in ALS (Pramatarova et al., 2001). Understanding the effect of ALS-specific mutations on astrocytes is paramount towards developing improved therapeutics.

Compared to neurons, astrocytes and other glial cells have been largely understudied (Barres, 2008). These cells play a crucial role in the maintenance of an extracellular environment that is suitable for neurons to function properly. Astrocytes remove neurotransmitters from the synapse, regulate local ion concentrations and pH homeostasis, supply glucose and other metabolites to neurons, remove metabolic waste, and aid in tissue repair after injury. They also help to regulate neuron excitability, synaptic strength, synaptogenesis, and the development of neuronal circuits (Reviewed by Clarke and Barres, 2013). Changes in normal astrocyte function can be toxic to neurons; it is becoming increasingly accepted that astrocytes play a key role in ALS and other neurodegenerative disorders.

Recent studies are revealing the complex genetic landscape underlying ALS risk, facilitating the development of improved animal models that more fully recapitulate key features of ALS pathology (Clement et al., 2003; Haidet-Phillips et al., 2013, p.; Qian et al., 2017). It remains to be seen whether inconsistent findings from these models is relevant to human disease or simply the shortcomings of mouse models. While some studies indicate that astrocytes expressing ALS-associated mutations can trigger disease onset, others show that selective expression of these mutations in astrocytes only impacts the rate of disease progression, and others suggest that astrocytes have no effect on disease pathology (Gong et al., 2000; Lino et al., 2002; Nagai et al., 2007; Qian et al., 2017; Wang et al., 2011; Yamanaka et al., 2008). Moreover, the complex genetic nature of sALS (which constitutes approximately 90% of ALS patients) makes it difficult to generalize the conclusions drawn from monogenic mouse models, especially given that many ALS-associated genes are differentially expressed in rodents (Oberheim et al., 2012, 2009; Zhang et al., 2016, 2014). It is important to develop additional models that better capture complex human genetics. Recent advances in human induced pluripotent stem cell (hiPSC) modeling provides an in vitro system to model ALS in patient-derived cells.

Here we consider the critical role that astrocytes play in ALS pathology. First, we establish the repertoire of ALS-associated genes that, due to their relatively high level of expression in astrocytes, indicate a role for astrocytes in disease pathogenesis. We discuss the mechanisms by which dysfunctional astrocytes contribute to neurodegeneration, with an emphasis on the ways in which hiPSC models have increased our understanding of the genetic underpinnings of astrocyte-related ALS pathology (Figure 1). Finally, we consider the implications for these findings in the context of other neurodegenerative diseases.

Figure 1: Investigating astrocyte involvement in neurodegeneration.

Figure 1:

Astrocytes can drive neurodegeneration through several interrelated mechanisms, including neuroinflammation, oxidative stress, excitotoxicity, and the promotion of protein aggregation in affected neurons. These pathological hallmarks of astrocyte-mediated neurodegeneration are conserved across a number of neurodegenerative diseases. ALS patients, for example, exhibit all facets of astrocyte-related disease pathology, and a large portion of ALS risk genes are highly expressed in astrocytes. However, the high degree of interrelation among astrocyte-related pathological phenotypes, as well as between astrocyte dysfunction and MN degeneration in ALS, begs the question of “which came first?” hiPSC models offer a new perspective for further examination of the “chicken-or-egg” dilemma in astrocyte-mediated neurodegeneration. Able to capture the complex, polygenetic landscape of ALS pathology, these models have already begun to yield new insight into the genetic and cellular underpinnings of neurodegeneration. However, hiPSC-derived astrocytes remain an under-used tool in examining non-cell autonomous disease pathology.

Genetic evidence of astrocyte involvement in ALS

Presently, approximately forty risk-associated genes have been identified in familial ALS, each of which is also mutated in a percentage of sporadic ALS cases (Chiò et al., 2012; Es et al., 2010; Li et al., 1988). The 5–10% of ALS patients with a family history of ALS are symptomatically indistinguishable from those presenting with sporadic ALS; 95% of all ALS patients develop TDP-43 aggregates. While ALS patients are genetically diverse, known ALS risk genes converge on several main cellular functions (Table 1, Figure 2) (Farg et al., 2013, p. 2; Hart and Gitler, 2012; Kim et al., 2010; Nihei et al., 2012). For example, ALS2, C21ORF2, C9ORF72, DCTN1, NEFH, NEK1, OPTN, PFN1, PRPH, TUBA4A, and VAPB are implicated in the maintenance of cytoskeletal dynamics/vesicular trafficking and release (Table 1). Many of the remainder are implicated in transcriptional regulation (ANG, FUS, SETX, SPG11, TARDBP), RNA metabolism/transport (ATXN2, C9ORF72, GLE1, HNRNPA1, HNRNPA2b1, MATR3, SMN1, TARDBP, TIA1), DNA repair (C21ORF2, NEK1, SPG11), protein trafficking/degradation (CCNF, CHMP2B, DCTN1, ERLIN2, FUS, SIGMAR1, UBQLN2, VCP), mitochondrial function (CHCHD10, SIGMAR1), endocytosis (ATXN2, CHMP2B), cell signaling (ALS2), and autophagy (SQSTM1, TARDBP, TBK1, VCP, OPTN). Expression of mutant genes can affect the relative abundance of other genes with similar cellular functions. For instance, expression of mSOD1(G93A) in astrocytes results in downregulation of Igf-1R (transcriptional regulation; pro-survival), and Rod1 (RNA metabolism/transport), and upregulation of Dcn1 (cytoskeletal dynamics/protein transport), Cx43 (cell signaling), and Nox2 (autophagy/endocytosis) (Almad et al., 2016; Marchetto et al., 2008; Marden et al., 2007; Vargas et al., 2008b). Still, the mechanisms by which mutations in these genes leads to the selective loss of motor neurons in an age-dependent manner is unclear.

Table 1: ALS risk-associated genes highly expressed in astrocytes.

Criteria for inclusion in table: FPKM for fetal/mature astrocytes greater than or equal to 10, unless gene was most highly expressed in fetal/mature astrocytes when compared to all brain cell-types (in which case, brain cell-types that showed similar gene expression levels were included). Genes for which hiPSC-derived astrocytes expressing disease-specific gene mutations have been generated and studied are written in red.

Gene Symbol Protein Human cell type specific expression [see (Zhang et al., 2014)] Normal protein function/Implicated pathways Number of disease-related mutations Possible pathologica l mechanism References
ALS2* Alsin (Amyotrophic lateral sclerosis 2) Fetal Astrocytes (FPKM >4, <8) Neurons (FPKM ~5.5) RAB5 GEF; Cytoskeletal maintenance; Vesicle and endosomal trafficking; Protein transport; Cell signaling; Glut4 trafficking ≥40 Endosomal dynamics; Oxidative stress (Zhang et al., 2014) (Chen et al., 2018) (Ince et al., 2011) (Zufiría et al., 2016) (Cirulli et al., 2015) (Morgan et al., 2015) (Sato et al., 2018)
ANG* Angiogenin Fetal Astrocytes (FPKM >0.75, <3.5) Microglia/Macropha ge (FPKM >1.5, <3) Ribonuclease; RNA transcription; Angiogenesis >10 Stress granule formation; TDP-43 aggregate formation (Zhang et al., 2014) (Chen et al., 2018) (Ince et al., 2011) (Zufiría et al., 2016) (Taylor et al., 2016)
ATXN2 Ataxin 2 Mature Astrocytes (FPKM >11, <20) Fetal Astrocytes (FPKM >11, <20) Neurons (FPKM ~10) Endocytosis; RNA metabolism CAG trinucleotide repeat expansion (27–33 repeats) TDP-43 toxicity modifier; Protein cleavage increases with length of polyglutamine repeat: truncated peptides potentially toxic to cells (Zhang et al., 2014) (Chen et al., 2018) (Zufiría et al., 2016) (Guerreiro et al., 2015) (Robberecht and Philips, 2013)
C21ORF2 C21ORF2 Fetal Astrocytes (FPKM >0.5, <2.5) Ciliome component; Possibly cytoskeletal dynamics; Possibly DNA repair ≥1 Mitochondrial dysfunction; Primary cilialoss; DNA repair insufficiency (Zhang et al., 2014) (Chen et al., 2018) (van Rheenen et al., 2016)
C9ORF72 C9ORF72 Microglia (FPKM >20, <30) Neurons (FPKM ~12) Fetal Astrocytes (FPKM >7, <15) RAB(8a/39b) GEF (experimentally); Possibly cytoskeletal dynamics and endosomal trafficking; Possibly lysosomal autophagy pathway; nucleocytoplasmic transport Intronic GGGGCC repeat (>30 repeats) (5 known Dipeptide Protein Repeat Sequences) Altered C9ORF72 RNA splicing; Toxic RNA or repeat dipeptide aggregation (due to reduced C9ORF72 expression); Ubiquitin and p62 aggregate formation; Neuro-inflammation; Formation of RNA foci; transcription inhibition; histone modification; accumulation of double stranded RNA (Zhang et al., 2014) (Chen et al., 2018) (Zufiría et al., 2016) (Taylor et al., 2016) (Guerreiro et al., 2015) (Almad et al., 2016) (Meyer et al., 2014) (Selvaraj et al., 2017) (Zhang et al., 2015) (Kwon et al., 2014) (Spang et al., 2014) (Gomez-Deza et al., 2015) (Allen et al., 2019) (Zhang et al., 2019)
CCNF Cyclin F Fetal Astrocytes (FPKM >0.6, <1.4) E3 ubiquitin-protein ligase complex component; Protein degradation ≥10 Abnormal ubiquitination; Ubiquitinated protein aggregation (including TDP-43 and SCFCyclin F substrate) (Zhang et al., 2014) (Chen et al., 2018) (Maurel et al., 2018) (Williams et al., 2016)
CHCHD10 Coiled-coil-helix-coiled-coil helix domain containing 10 Mature Astrocytes (FPKM >1.75, <3.5) Mitochondrial protein of unknown function ≥2 Mitochondrial function (Zhang et al., 2014) (Chen et al., 2018) (Taylor et al., 2016) (Guerreiro et al., 2015) (Ajroud-Driss et al., 2015)(Perron e et al., 2017)(Che et al., 2017)
DCTN1 Dyactin subunit p150glued Fetal Astrocytes (FPKM >3.5, <8) Oligodendrocytes (FPKM >2.3, <3.2) Neurons (FPKM ~2.5) Mature Astrocytes (FPKM >1.5, <2.25) Dynein motor complex component; Vesicle trafficking; Cytoskeletal dynamics ≥10 Protein transport (Zhang et al., 2014) (Chen et al., 2018) (Taylor et al., 2016) (Guerreiro et al., 2015) (Boillée et al., 2006)
ERLIN2 ER lipid raft associated 2 Mature Astrocytes (FPKM >9, <12) ER lipid formation raft ≥1 Lipid raft functioning (Zhang et al., 2014) (Guerreiro et al., 2015)
FUS FUS Fetal Astrocytes (FPKM >6, <8) Microglia/Macrophage (FPKM <4.5, >7) Transcriptional regulator; UPS pathway ≥35 Altered RNA processing resulting in FUS aggregates (Zhang et al., 2014) (Chen et al., 2018) (Zufiría et al., 2016) (Taylor et al., 2016) (Guerreiro et al., 2015) (Maurel et al., 2018)
GLE1 GLE1 RNA export mediator Fetal Astrocytes (FPKM >1.75, <3.5) Mature Astrocytes (FPKM >2, <2.5) Neurons (FPKM~2.2) RNA metabolism ≥3 RNA metabolism (Zhang et al., 2014) (Zufiría et al., 2016) (Guerreiro et al., 2015) (Kaneb et al., 2015)
GLT8D1 Glycosyltransferase 8 domain containing 1 Neurons (FPKM >12) Mature Astrocytes (FPKM >9, <12) Unknown; Possibly involved in sugar metabolism and cell adhesion 5 Unknown (Cooper- Knock et al., 2019; Zhang et al., 2014)
HNRNPA1 Heterogeneous nuclear ribonucleoprotein A1 Fetal Astrocytes (FPKM >70, <140) Oligodendrocytes (FPKM >80, <115) Microglia/Macropha ge (FPKM >70, <90) Neurons (FPKM~70) Mature Astrocytes (FPKM >25, <40) RNA-binding protein; Directly interacts with TDP-43; RNA metabolism; transport of poly(A) mRNA from nucleus to cytoplasm 3 Strengthened ‘stericzipper’ motifin PrLD; Accelerated formation of fibrils that self-seed and cross-seed polymerize with wildtype hnRNP (Zhang et al., 2014) (Chen et al., 2018) (Zufiría et al., 2016) (Taylor et al., 2016) (Guerreiro et al., 2015) (Hutten and Dormann, 2016)
HNRNPA2b1 Heterogeneous nuclear ribonucleoprotein A2/B1 Fetal Astrocytes (FPKM >60, <100) Microglia/Macrophage (FPKM >60, <90) Neurons (FPKM~65) Oligodendrocytes (FPKM >50, <80) Mature Astrocytes (FPKM >40, <50) RNA-binding protein; Directly interacts with TDP-43; RNA metabolism ≥1 Strengthened ‘stericzipper’ motifin PrLD; Accelerated formation of fibrils that self-seed and cross-seed polymerize with whnRNP; Widespread changes in alternative splicing (mechanism unknown) (Zhang et al., 2014) (Chen et al., 2018) (Zufiría et al., 2016) (Guerreiro et al., 2015) (Kaneb et al., 2015)
MATR3 Matrin 3 Neurons (FPKM~75) Fetal Astrocytes (FPKM >35, <50) Oligodendrocytes (FPKM >25, <40) Microglia/Macrophage (FPKM >25, <35) Mature Astrocytes (FPKM >15, <20) RNA-binding protein; RNA metabolism 4 Interaction with TDP-43 to regulate transcription (in MNs) (Zhang et al., 2014) (Chen et al., 2018) (Zufiría et al., 2016) (Taylor et al., 2016)
NEK1 Never-in-mitosis A (NIMA-) related kinase 1 Mature Astrocytes (FPKM >5, <7) Fetal Astrocytes (>3, <4) Centrosomal complex component; Mitotic protein kinase; Cytoskeletal dynamics; Genotoxic stress response ≥120 DNA damage accumulation (Zhang et al., 2014) (Chen et al., 2018) (Higelin et al., 2018) (Chen et al., 2011) (Pelegrini et al., 2010) (Kenna et al., 2016)
PFN1 Profilin-1 Microglia/Macropha ge (FPKM >25, <65) Fetal Astrocytes (FPKM >25, <40) Oligodendrocytes (FPKM >15, <25) Mature Astrocytes (>12, 20) Actin-binding protein; Cytoskeletal dynamics; regulator of actin polymerization 5 Cytoskeletal pathways disruption (Zhang et al., 2014) (Chen et al., 2018) (Zufiría et al., 2016) (Taylor et al., 2016)
SIGMAR1 Sigma1R Fetal Astrocytes (FPKM >6, <9) Mature Astrocytes (FPKM >5, <7) Neurons (FPKM ~6) Regulator of Ca2+ transport from ER to mitochondria through IP3R; Protein degradation; receptor protein involved in endocrine and immune signaling ≥1 Aberrant potassium channel (Kv1.4) activity; Mitochondri a-associated membrane collapse (Zhang et al., 2014) (Chen et al., 2018) (Zufiría et al., 2016) (Watanabe et al., 2016) (Shinoda et al., 2015)
SPG11 Spatacsin Mature Astrocytes (FPKM >6.5, <8) Fetal Astrocytes (FPKM >4.5, <6.5) Microglia/Macrophage (FPKM >5.25, <6.5) Oligodendrocytes (FPKM >5, <6) Neurons (FPKM ~5.3) Possibly implicated in gene expression control, protein transport, and/or DNA damage repair ≥21 DNA damage repair (Zhang et al., 2014) (Chen et al., 2018) (Morgan et al., 2015) (Nakamura et al., 2016)
SOD1 Cu/Zn superoxide dismutase Neurons (FPKM~190) Mature Astrocytes (FPKM >100, <125) Oligodendrocytes (FPKM >75, <115) Fetal Astrocytes (FPKM >25, <65) Microglia/Macropha ge (FPKM >20, <35) Nuclear/cytosolic protein; Superoxide radical break-down; UPS/chaperone network >200 SOD1 and p62 protein/RNA aggregate formation; Gain in redox function; Axonal transport (Zhang et al., 2014) (Chen et al., 2018) (Zufiría et al., 2016) (Taylor et al., 2016) (Guerreiro et al., 2015) (Almad et al., 2016) (Meyer et al., 2014) (Maurel et al., 2018)
SQSTM1 p62 Microglia/Macropha ge (FPKM >15, >30) Mature Astrocytes (FPKM >15, <20) Oligodendrocytes (FPKM >10, <20) Neurons (FPKM ~18) Autophagy adaptor; Glucose metabolism regulation; ERAD pathway; NF-kB pathway ≥10 Autophagy (Zhang et al., 2014) (Chen et al., 2018) (Zufiría et al., 2016) (Taylor et al., 2016) (Guerreiro et al., 2015) (Maurel et al., 2018)
TARDBP TAR DNA-binding protein Neurons (FPKM~32) Fetal Astrocytes (FPKM >17, <30) Oligodendrocytes (FPKM >25, <30) Microglia (FPKM >20, <25) Mature Astrocytes (FPKM >15, <18) RNA-binding protein; RNA splicing; Autophagy; transcriptional regulator ≥60 DNA/RNA metabolic changes; Ubiquitin, p62, and TDP-43 aggregate formation (Zhang et al., 2014) (Chen et al., 2018) (Zufiría et al., 2016) (Taylor et al., 2016) (Guerreiro et al., 2015) (Maurel et al., 2018) (Serio and Patani, 2018)
TIA1 T-cell restricted intracellularantigen 1 Fetal Astrocytes (FPKM >14, <20) Neurons (FPKM~12) RNA binding protein; Stress granule (SG) component ≥1 Enhanced phase separation of membrane-less organelles; Delayed SG disassembly ; TDP-43 aggregate formation (Zhang et al., 2014) (Chen et al., 2018) (Mackenzie et al., 2017)
VAPB VAMP-associated membrane protein B Fetal Astrocytes (FPKM >0.15, <0.32) Vesicular trafficking; Unfolded protein response pathway 4 Altered binding to mitochondrial proteins; Elevated mitochondrial calcium; TDP43 aggregate formation (Zhang et al., 2014) (Zufiría et al., 2016) (Maurel et al., 2018) (Boillée et al., 2006) (Kim et al., 2016) (Tsuda et al., 2008)
VCP Valosin-containing protein (Transitional endoplasmic reticulum ATPase) FetalAstrocytes (FPKM >18, <14) Neurons (FPKM ~10) Microglia/Macrophage (FPKM >6, <14) Ubiquitin segregase; Protein degradation; UPS/autophagy/ERAD pathways 5 Autophagy; Proteasomal degradation; TDP-43 aggregate formation (Zhang et al., 2014) (Chen et al., 2018) (Zufiría et al., 2016) (Taylor et al., 2016) (Guerreiro et al., 2015) (Maurel et al., 2018) (Hall et al., 2017)

Figure 2: Mechanisms implicating astrocytes in ALS-related neurodegeneration.

Figure 2:

(1) Patient astrocytes show enhanced reactivity and heightened neuroinflammation, with astrocytes favoring pro-over anti-inflammatory reactive phenotypes. In addition to being directly toxic to MNs, neuroinflammatory factors promote cytotoxic protein aggregation (i.e. via TGF-Beta) and dysregulate astrocytic control of synaptic glutamate levels (i.e. via TNF-alpha). (2) By generating a number of reactive oxygen and nitrogen species (black dots), diseased astrocytes provoke oxidative stress, which can directly kill MNs via the oxidation of key macromolecules. Oxidative species can also promote further neurodegeneration by disrupting mitochondrial functions (in both astrocytes and MNs) and enhancing astrocytic neuroinflammation. Reactive oxygen species can also drive pathological protein aggregation in MNs and lower MN defenses against excitotoxic death. (3) Due to enhanced Cx-43 expression and increased vesicular release of glutamate-containing vesicles, patient astrocytes show heightened glutamate release (purple dots) (3-a). Together with a decrease in glutamate re-uptake (3-b), this drives the build-up of glutamate at the synapse. The effects of glutamate accumulation are subsequently compounded by the astrocyte-driven enhancement of Ca2+ (blue dots) conductance in MNs (3-c). (4) Astrocytes also incite protein aggregation in MNs by inhibiting MN autophagic capacity and by secreting prion-like protein aggregates.

RNAseq data from human neural cell subtypes indicate that many of the genes that have been associated with ALS are expressed in astrocytes at similar or higher levels than in neurons (Table 1) (Zhang et al., 2014). While the majority of previous work examining these genes has focused on MNs, given the range of essential cellular functions that these mutated proteins perform, it seems likely that astrocyte function is also impacted. Astrocyte dysfunction may affect the onset and/or progression of neurodegeneration via several possible, non-mutually exclusive mechanisms: the increased production of neurotoxic inflammatory factors and reactive oxygen species (ROS), the disruption of astrocyte-mediated MN homeostasis, the establishment of astrocyte-mediated excitotoxic conditions coupled with enhanced neuronal susceptibility to excitotoxic death, and the accumulation of pathological protein aggregates (Figure 2).

Astrocyte dysfunction in ALS

The role of astrocytes in ALS onset and progression remains unclear. Early models suggested that astrocytic dysfunction was secondary to neuronal death (Alexianu et al., 2001; Cassina et al., 2008; Simpson et al., 2004; West et al., 2004; Wilms et al., 2010; Yoshihara et al., 2002). The onset of neurodegeneration was thought to arise from neuron cell-autonomous mechanisms, with astrocytes only implicated in further disease progression. Consistent with this, specific removal of mutant SOD1 from astrocytes in the SOD1(G37R) ALS mouse model slows disease progression, but does not impact disease onset (Yamanaka et al., 2008). In contrast, deletion of mutant SOD1 from astrocytes in the SOD1(G85R) mouse delays disease onset and progression (Lino et al., 2002; Wang et al., 2011). Moreover, chimeric mouse studies using SOD1(G93A) demonstrated that neighboring wildtype glia extend the survival of SOD1 mutant (mSOD1) MNs (Clement et al., 2003). These observations are supported by in vitro studies of astrocytes expressing disease-specific mutations in SOD1—including G93A (Di Giorgio et al., 2007; Nagai et al., 2007), A4V (Haidet-Phillips et al., 2011), and G37R (Marchetto et al., 2008)—as well as astrocytes from sporadic ALS (sALS) patients (Haidet-Phillips et al., 2011; Qian et al., 2017). Taken together, these findings suggest that MN death may be initiated in a non-cell autonomous astrocyte-dependent manner. Similar inconsistencies regarding astrocytic involvement in disease pathogenesis have also been noted upon examination of other ALS-associated genes—including TARDBP and C9ORF72 (Allen et al., 2019; Haidet-Phillips et al., 2013; Koppers et al., 2015; Lagier-Tourenne et al., 2013; Meyer et al., 2014; Tong et al., 2013; Waite et al., 2014). The disparity between these results may be attributable to differences between models (e.g. promotors used to drive mutant gene expression, the use of different disease-associated mutants of the same gene, etc.). More work must be done in order to understand the extent to which astrocytes can be an initial driver of ALS.

Given that astrocyte dysfunction is evident in ALS pathology, understanding the mechanisms by which astrocytes contribute to disease could have major implications for potential drug targets. Importantly, these mechanisms are not isolated from each other, but rather are highly interconnected, often amplifying one another through a series of positive feedback loops. For example, the initiation of the inflammatory response results in the release of ROS from astrocytes, which in turn induces further reactivity in astrocytes, acts as a signaling molecule that affects neuronal susceptibility to excitotoxic stressors, and induces further ROS release from degenerating MNs and newly activated glial cells (Figure 1). The highly responsive nature of the positive feedback loops implicated in astrocyte-driven neurodegeneration has made it difficult to distinguish between the initiating and progressive causes of neurodegeneration.

The utility of hiPSCs in examining astrocyte-related ALS pathology

Murine ALS models have significantly enhanced our understanding of several risk-associated mutations, but fail to capture the complexity of human genetics. Specifically, the structural and functional differences between human and rodent astrocytes make it difficult to generalize conclusions pertaining to astrocyte-related pathology that are drawn from rodent models. Human astrocytes, for example, are larger and have 10-fold more glial fibrillary acidic protein (GFAP)-positive primary processes than astrocytes found in rodents (Oberheim et al., 2009). Unlike rodent astrocytes, human astrocytes respond to glutamatergic and purinergic receptor agonists with a transient increase in cytosolic Ca2+. Rodent astrocytes are also less structurally and functionally diverse than human astrocytes (Oberheim et al., 2012, 2009). This is a significant detriment to rodent models, as this diversity may explain the highly localized nature of neuronal death that is observed in many neurodegenerative diseases (reviewed by Schitine et al., 2015). Finally, in the context of ALS, it is important to note that human and rodent astrocytes also differ in the relative expression of several ALS-related genes when compared with neurons (Oberheim et al., 2012, 2009; Zhang et al., 2016, 2014). When combined with CRISPR-mediated genome editing, patient-derived hiPSCs represent a powerful tool for assessing the impact of ALS-specific mutations in varied genetic backgrounds. Patient-derived hiPSCs can also be used to study those sporadic ALS cases in which no known mutations have yet been identified. Thus, hiPSCs represent a valuable tool in the study of astrocyte-related ALS pathology.

Many protocols have been established for the generation of astrocytes from patient-derived hiPSCs and neural progenitor cells (NPCs) (Emdad et al., 2011; Santos et al., 2017; Shaltouki Atossa et al., 2013; Tcw et al., 2017). The earliest protocols, which took as long as 180 days, generated relatively uniform populations of astrocytes by progressing through neuroepithelial cell intermediates that were patterned with anterior/posterior/ventral (fibroblast growth factor 8/retinoic acid/sonic hedgehog) patterning molecules (Emdad et al., 2011; Krencik et al., 2011). Conversely, more recent protocols yield functional astrocytes in as little as 30 days, though these methods are as yet unable to produce pure populations of distinct astroglial subtypes (Shaltouki Atossa et al., 2013; Tcw et al., 2017). Most recently, via the overexpression of two transcription factors (SOX9 and NFIB), astrocytes can be generated in as little as 2 weeks (Canals et al., 2018). hiPSC-derived astrocytes display several astrocyte-specific and disease-relevant functionalities, such as phagocytic ability and neuroinflammatory response (Tcw et al., 2017; Santos et al., 2017).

To date, hiPSCs have been generated from patients with a variety of mutations in SOD1 (Bhinge et al., 2017; Chen et al., 2014; Jeon et al., 2018; Kiskinis et al., 2014; Li et al., 2015; Wainger et al., 2014; Wang et al., 2017), C9ORF72 (Donnelly et al., 2013; Egawa et al., 2012; Haeusler et al., 2014; Li et al., 2015; Liu et al., 2017; Madill et al., 2017; Meyer et al., 2014; Sareen et al., 2013; Wen et al., 2014; Westergard et al., 2016), TARDBP (Alami et al., 2014; Barmada et al., 2014; Bilican et al., 2012; Serio et al., 2013; Zhang et al., 2013), FUS (Ichiyanagi et al., 2016; Japtok et al., 2015; Lenzi et al., 2016, 2015; Li et al., 2015; Wainger et al., 2014; Wang et al., 2017), ANG (Li et al., 2015), FIG4 (Li et al., 2015), VAPB (Mitne-Neto et al., 2011), and VCP (Hall et al., 2017; Li et al., 2015). A smaller number of these hiPSCs have yet been differentiated into astrocytes: mSOD1 (Almad et al., 2016; Tyzack et al., 2017), mC9ORF72 (Donnelly et al., 2013; Liu et al., 2017; Madill et al., 2017; Meyer et al., 2014), mTARDBP (Barmada et al., 2014; Serio et al., 2013), FUS (Qosa et al., 2016), and VCP (Hall et al., 2017). The results of these studies, which will be discussed throughout the following sections, have already begun to inform our understanding of astrocyte-related processes in ALS.

Critically, as with other hiPSC-derived cell-types, hiPSC-derived astrocytes display fetal transcriptional profiles (Reviewed by Ravaioli et al., 2018). This complicates the application of hiPSCs for the study of age-related disorders such as ALS. While methods to address this issue do exist, they are limited in their ability to properly recapitulate the normal aging process. For example, progerin-overexpression induces some age-related phenotypes, but does not facilitate maturation of neural cell types (Miller et al., 2013). While extended culture can improve the maturity of hiPSC-derived astrocytes, even organoid-based differentiations of up to a year only approach neonatal transcriptomic profiles (Sloan et al., 2017). Alternatively, transcription-factor induction from fibroblasts maintains epigenetic age, and have been applied to studies of MNs in ALS (Liu et al., 2016; Son et al., 2011; Q.-J. Zhang et al., 2017); similar protocols for astrocyte induction have been reported but not yet applied to ALS (Caiazzo et al., 2014; Tian et al., 2016). Nonetheless, hiPSC models well capture the genetic elements of disease risk (if not the disease state itself) and yield useful insight into the origins of ALS.

The neurotoxic effects of neuroinflammation

In healthy brain tissue, neuroinflammation occurs as part of a protective homeostatic reaction to acute states. For example, the activation of the immune response following an infection or injury can trigger the onset of neuroinflammation (reviewed by Dantzer et al., 2008 & Tohidpour et al., 2017). This communication between the peripheral immune system and the brain is crucial for the induction of normal, adaptive responses to infection or injury (e.g. fever, fatigue, etc.) that are necessary for recovery (Serrats et al., 2010; Hansen et al., 2001; Laflamme et al., 1999; Ching et al., 2007). Moreover, neuroinflammation coincides with the onset of tissue repair responses. However, the neuroinflammation that results from immune activity – characterized by the short-lived activation of both astrocytes and microglia – is highly-regulated and transient (Norden et al., 2016).

Neuroinflammation can be identified by increased abundance of pro-inflammatory biomarkers, such as cytokines (IL-1β, IL-6, and TNF-α), chemokines (CCL2, CCL5, and CXCL1), and secondary messengers such as Nitrous Oxide (NO), prostaglandins, and ROS, in CNS tissue (Chao et al., 1996; Meeuwsen et al., 2003). At the cellular level, the initiation, maintenance, and regulation of neuro-immune responses is accomplished by activated microglia and astrocytes, each contributing to distinct and complementary aspects of inflammation. The development of reactive gliosis – the accumulation of the morphologically and functionally altered glial cells that are involved in the neuro-immune response – is a key feature of neuroinflammation. During an acute neuro-immune response, microglia can adopt an “M1” pro-inflammatory phenotype or “M2” protective/anti-inflammatory function depending on the type of insult (Reviewed by Cherry et al., 2014). Similarly, reactive astrocytes can both contribute to (“A1”) and prevent (“A2”) damage from inflammation, and the specifics of destructive and protective reactive astrocytes will be discussed presently (Brambilla et al., 2005; Colombo et al., 2012; Hindinger et al., 2012; Linker et al., 2010; Nobuta et al., 2012).

Neuroinflammation contributes to neurodegeneration

Despite its contribution to cell repair mechanisms, rampant neuroinflammation is observed in several neurodegenerative diseases, including Alzheimer’s disease (AD), multiple sclerosis (MS), Parkinson’s disease (PD) (Ellwardt and Zipp, 2014; Heneka et al., 2015; Tansey and Goldberg, 2010), and ALS (Morello et al., 2017). Such neuroinflammation is not evident in asymptomatic individuals carrying ALS-associated mutations, demonstrating that it is a characteristic that is necessarily linked to disease development (Oeckl et al., 2018). Damaging pro-neuroinflammatory molecules, such as NO and ROS, can indirectly induce MN degeneration via activation of intracellular signaling cascades that alter key cellular functions (Pintado et al., 2017). These findings suggest that alterations to the regulated balance between the inflammatory and protective components of neuroinflammation can result in a cytotoxic microenvironment and subsequent neurodegeneration (Chao et al., 1996; Downen et al., 1999; McCoy et al., 2006).

A prominent hypothesis regarding the role of neuroinflammation in neurodegeneration is that neurotoxic inflammation results, in part, from a high ratio of M1/M2 reactive microglial populations—a trait that occurs even in normal aging (Frank et al., 2006; Kigerl et al., 2009; Kumar et al., 2013; F. Zhang et al., 2017). Astrocytes display a similar imbalance between cellular phenotypes in disease pathology (Liddelow et al., 2017). Consequently, deactivation of the pro-inflammatory astrocyte signaling cascade (NF-kB inhibition) minimizes the extent of tissue injury following an inflammatory response (Brambilla et al., 2012, 2009, 2005). However, it remains unclear whether this aberrant inflammatory response is self-generated or initiated following neuronal damage of unknown origin. Regardless, it is clear that neurodegeneration and inflammation promote each other through a positive feedback loop: inflammatory cytokines induce neurodegeneration, while MN decay incites further inflammation (Figure 1) (Meeuwsen et al., 2003). This feedback mechanism may contribute to the progressive nature of ALS. However, the presence of reactive astrocytes and microglia before the onset of motor symptoms in mouse models of ALS suggests that astrocyte dysfunction plays a more inciting role in disease pathology (Bruijn et al., 1997; Levine et al., 1999). For example, astrocytes can induce neuroinflammation through the release of pro-inflammatory factors that increase blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB) permeability, indicating a potential mechanism for astrocyte involvement in disease onset (Abbott, 2000; Winkler et al., 2014).

Toxic gain-of-function mutations contribute to neuroinflammation in ALS

Several ALS-associated genes that are expressed in astrocytes are implicated in hyperactive neuroinflammatory responses in ALS patients. Astrocytes expressing mSOD1, for instance, display an enhanced release of neurodegenerative inflammatory factors and pro-inflammatory mediators (Chao et al., 1996; Dawson et al., 1993; Gong et al., 2000; Meeuwsen et al., 2003; Pintado et al., 2017). Interestingly, gliosis is suppressed when autophagy is conditionally knocked out in MNs in the SOD1 G93A mouse model (Maniatis et al., 2019; Rudnick et al., 2017), suggesting a possible mechanism for MNs to promote inflammation in neighboring glia. SOD1 mutations attenuate neuronal responses to protective aspects of inflammation through altered TGF-β signaling, which is observed in SOD1(G93A) mice (Phatnani et al., 2013) as well as in ALS patients (Endo et al., 2015). Prolonged release of TGF-β1 from chronically reactive astrocytes (both wildtype and ALS-related SOD1 mutant) drives the development of ALS-like protein inclusions in vitro and induces cell death in otherwise healthy MNs (Tripathi et al., 2017). Furthermore, reactive murine astrocytes harboring the SOD1(G93A) mutation release increased levels of nerve-growth factor (NGF) (Ferraiuolo et al., 2011). Although NGF is neuro-protective in non-pathological conditions, its low-affinity binding to p75 neurotrophin receptors (p75NTR) induces apoptosis. Interestingly, aberrant NGF processing in mSOD1 astrocytes leads to p75NTR upregulation in nearby neurons, an outcome that is also noted in spinal MNs from ALS patients (Ferraiuolo et al., 2011). Subsequent apoptosis can be prevented by application of NGF or p75NTR blocking-antibodies (Pehar Mariana et al., 2004; Turner Bradley J. et al., 2004, p. 7). Still, these toxic effects of SOD1 mutations are not ubiquitous—not all SOD1 mutations, when selectively expressed in astrocytes, are enough to drive MN degeneration (Gong et al., 2000; Nagai et al., 2007). Finally, by using hiPSC-derived astrocytes harboring the SOD1(A4V) mutation to model intercellular interactions at the BBB, it has been demonstrated that patient astrocyte dysfunction may induce alterations to the BBB (Qosa et al., 2016). Interestingly, hiPSC-derived astrocytes expressing FUS(H517Q) induced similar effects (Qosa et al., 2016).

Selective expression of mutant TDP-43 (Q331K) in glial cells promotes an increased level of age-dependent astrocyte activation (Ditsworth et al., 2017), but the mechanism for this enhanced neuro-immune response is unclear. TDP-43 is involved in transcriptional regulation and RNA metabolism, and one explanation is that TDP-43 mutants favor expression of pro-inflammatory genes (Table 1) (Reviewed by Lagier-Tourenne et al., 2010; Swarup et al., 2011). Lipocalin 2 (Lcn2) protein is one such pro-inflammatory factor that is secreted by reactive astrocytes expressing the patient-specific TDP-43(M337V) mutant (Tong et al., 2013), is toxic to neurons in a non-specific manner (Song et al., 2014), and linked to neuro-inflammatory response (Jin et al., 2014). Lcn2 also promotes permeabilization of the BBB (Egashira et al., 2016). However, Lcn2 secretion from reactive astrocytes has, thus far, only been observed in rodent models (Lee et al., 2009). Given that hiPSC-derived astrocytes harboring TARDBP mutations do not induce MN degeneration, future studies should examine whether LCN2 secretion is observed from mTARDBP+ patient-derived astrocytes (Serio et al., 2013).

The C9ORF72 hexanucleotide repeat expansion—the most pervasive risk-associated gene in ALS pathology—has also been implicated in the dysregulated neuroinflammation seen in ALS patients (DeJesus-Hernandez et al., 2011; Renton et al., 2011). C9ORF72 is thought to play a role in endosomal trafficking and the lysosomal autophagy pathway (Table 1). It acts as a guanidine nucleotide exchange factor (GEF) for Rab8, which is also highly expressed in astrocytes (fetal) (Sellier et al., 2016; Zhang et al., 2014) and involved in protective, non-inflammatory signaling (Wall et al., 2017). In general, C9ORF72 expansions may promote neurodegeneration through two possible mechanisms: 1) haploinsufficiency of the C9ORF72 gene (loss-of-function, LOF) (DeJesus-Hernandez et al., 2011; Waite et al., 2014), and/or 2) toxicity of transcribed and/or translated elements of the mutant intronic region of the gene (GOF) (Reviewed by La Spada and Taylor, 2010). Unlike GOF phenotypes that can directly induce neurotoxicity, C9ORF72 LOF appears to be more relevant to the neuroinflammatory component of ALS, gradually altering the extracellular environment over the course of an animal’s lifetime. For example, complete KO of C9ORF72 in mice results in heightened age-related inflammation, including neuroinflammation (Atanasio et al., 2016; Koppers et al., 2015; O’Rourke et al., 2016). The widening gap between patients’ and controls’ neuroinflammatory responses with age may be relevant in considering the age-related nature of ALS pathology.

The exact role neuroinflammation plays in the pathogenesis of ALS remains unclear even as evidence of its involvement increases. Neuroinflammation may be a byproduct of tissue damage from the disease, or may even be protective, attempting to promote tissue repair early in disease progression (Endo et al., 2015; Hu et al., 2012). The bifunctional roles of neuro-immune cells may well explain the emergence of such diametrically contrasting conclusions, and it is possible that reactive astrocytes play dual roles in disease pathology given the differences in function between inflammatory A1 and protective A2 astrocyte populations. The increased presence of A1 astrocytes in post-mortem CNS tissue in ALS (Liddelow et al., 2017) is consistent with neuroinflammation that could lead to MN death. As the disease progresses, the relative decrease in cells displaying A2 protective phenotypes compounds the directly neurotoxic effects of A1 cells.

Experiments using hiPSC-derived cells indicates that specific ALS-related mutations can either block protective A1 phenotypes or generate toxic A2 phenotypes. For example, upregulation of EphB1 in degenerating MNs induces astrocytes to take on protective phenotypes (Tyzack et al., 2017). Disruptions in the EphB1-ephrin-B1 pathway in hiPSC-derived astrocytes from patients harboring SOD1(D90A) mutations result in a relative decrease in the number of reactive astrocytes with neuroprotective phenotypes. In accordance with this, hiPSC-derived astrocytes from sporadic ALS patients transplanted into mice display neuroinflammatory reactive phenotypes and are toxic to surrounding neurons, whereas control astrocytes do not (Qian et al., 2017). However, because the resultant neurodegeneration was ubiquitous, this did not explain the selective toxicity observed in MN populations. Thus, future work using hiPSC-derived astrocytes should focus on protocols that generate astrocytes that closely mimic the A1 and A2 phenotypes observed in vivo, and whether mutations implicated in ALS fundamentally alter the function of these distinct astrocyte populations.

The generation and proliferation of oxidative stress

Endogenous ROS are generally a byproduct of mitochondrial enzymes (Cadenas et al., 1977). In small amounts, free radicals serve as important signaling molecules in the establishment of long term potentiation (LTP), the myelination of highly active neurons, and angiogenesis in damaged brain tissue (Atkins et al., 1997; Atkins and Sweatt, 1999; McCann and Roulston, 2013; Wang et al., 2004; Zimmerman et al., 2005). In large quantities, however, ROS are cytotoxic – capable of oxidizing proteins, DNA, and other essential molecules. ROS contribute to neurodegeneration by driving inflammation, pathological protein aggregation, mitochondrial dysfunction, activation of apoptotic pathways, and disruption the BBB (Figure 2) (Cassina et al., 2008; Cohen et al., 2012; Haorah et al., 2007; Javed et al., 2012; Ryter et al., 2007; Shodai et al., 2013). Finally, by reacting with pro-inflammatory NO, which increases as part of the hyper-active neuroinflammatory response, ROS can be converted into cytotoxic reactive nitrogen species (RNS) (Kempuraj et al., 2016). Thus, the balance between the production and breakdown of ROS is crucial.

ROS are associated with aging and neurodegeneration

Oxidative damage is a nearly ubiquitous feature in post mortem brain and spinal cord samples from ALS patients (P. J. Shaw et al., 1995; Shibata et al., 2001). Accordingly, disease models have demonstrated that ROS increase with age and are direct drivers of MN death (Cassina et al., 2008; Cohen et al., 2012; Haorah et al., 2007; Javed et al., 2012; Ryter et al., 2007; Shodai et al., 2013). DNA damage resulting from oxidative stress, as measured by 8-hydroxy-2′-deoxyguanosine (8-OHdG) accumulation, has been observed in postmortem spinal cord and motor cortex from ALS patients (Ferrante et al., 1997). Moreover, 8-OHdG accumulation in cerebrospinal fluid and urine increases during the course of disease. It has been proposed that DNA damage contributes to fALS and sALS pathogenesis (Reviewed by Coppedè and Migliore, 2015). Indeed, alterations to DNA damage response signaling pathways causes neurodegeneration in hiPSC-derived motor neurons (Naumann et al., 2018). However, whether the increase in ROS occurs before disease onset or is secondary to initial MN degeneration remains uncertain. It is important to note that ROS-induced mitochondrial dysfunction promotes further ROS production, initiating a positive feedback loop, suggesting that the accumulation of ROS contributes to the progressive nature of ALS (Figure 2).

Astrocyte Dysfunction Contributes to ROS Build-up

Paradoxically, reactive astrocytes contribute to the production of ROS while also scavenging ROS (Schreiner et al., 2015), suggesting that a balance between protective and inflammatory reactive astrocyte phenotypes may be crucial for the maintenance of a neuron-friendly extracellular environment. A deeper investigation into how astrocytes contribute to the normal age-related increase in ROS production, and how this differs between patients and controls, may offer further insight into how and why ROS increase with aging and in ALS pathology. Present theories mainly highlight mitochondrial dysfunction in patient cells as a potential explanation for the disease-related increase in ROS production. Interestingly, much like ROS production, mitochondrial dysfunction is progressively enhanced over the course of normal aging, and is exacerbated in ALS and other neurodegenerative diseases (Cassina et al., 2008; Corral-Debrinski et al., 1992; Johri and Beal, 2012; Said Ahmed et al., 2000). Drug-induced mitochondrial disruption in wildtype astrocytes incites MN degeneration upon co-culture (Cassina et al., 2008), and the stimulation of antioxidant pathways in astrocytes delays symptomatic onset, and extends survival in animal models (Vargas et al., 2008a). Further examination of these pathways in hiPSC-astrocytes may clarify whether patient lines inherently produce more ROS and/or are more prone to DNA damage, as well as the impact patient astrocytes have on motor neuron function.

Risk-Associated Genes and Astrocyte Contribution to ROS Accumulation

Several ALS-associated genes have been implicated in disease-related mitochondrial dysfunction. For example, the SOD1 enzyme, which typically functions as a potent antioxidant (Reviewed by Kim, 2014), aberrantly accumulates in the mitochondria of neural cells from ALS patients and murine models. The extent of such SOD1 accumulation increases with age, and incites mitochondrial dysfunction and increased ROS production (Liu et al., 2004; Takeuchi et al., 2002; Velde et al., 2008; Zetterström et al., 2007). Interestingly, an over-oxidized form of wildtype SOD1, which damages mitochondria and is prone to aggregation, has been observed in a subset of sporadic ALS patients (Guareschi et al., 2012). Thus, post-translational modifications may drive aggregation of SOD1 (Bosco et al., 2010; Ezzi et al., 2007; Guareschi et al., 2012; Schmitt and Agar, 2017), indicating that the localization and/or oxidation state of SOD1 may incite mitochondrial dysfunction (Said Ahmed et al., 2000; Xu et al., 2018).

Other ALS-related genes code for proteins that are critical for proper mitochondrial function (C21ORF2, CHCHD10, SIGMAR1, and VAPB - Table 1). CHCHD10 is a key regulator of potentially ROS-generating mitochondrial oxidative phosphorylation reactions (Purandare et al., 2018). The endogenous expression or forced overexpression of mutant CHCHD10 (mCHCHD10) (S59L) in HeLa cells and patient fibroblasts disrupts mitochondrial structure (Bannwarth et al., 2014; Genin et al., 2016). CHCHD10 disease variants (G66V and P80L) also display faulty interactions with cytochrome c oxidase and other mitochondrial proteins, resulting in reduced respiration and increased ROS. To the same effect, mutations in SIGMAR1 disrupt mitochondrial function, leading to an increase in ROS production and autophagic cell death (Fukunaga et al., 2015; Shinoda et al., 2015; Watanabe et al., 2016). As a receptor that is enriched at the mitochondrial-associated endoplasmic reticulum membrane, SIGMAR1 is important for mitochondrial homeostasis (Hayashi and Su, 2007; Pal et al., 2012). VAPB, which is also enriched at the mitochondrial-associated endoplasmic reticulum membrane, is involved in mitochondrial Ca2+ uptake (De Vos et al., 2012). The expression of ALS mutant VAPB (P56S) results in mitochondrial damage. Importantly, the relatively high expression of SIGMAR1 and VAPB in fetal astrocytes suggests that further study of mutations in these genes may shed light on the developmental underpinnings of mitochondrial dysfunction in ALS. Finally, although not mitochondrial proteins, the mTDP-43 and C9ORF72 repeat has also been associated with mitochondrial dysfunction and the upregulation of oxidative stress markers (Braun et al., 2011; Duan et al., 2010; Lopez-Gonzalez et al., 2016; Zhan et al., 2015).

In hiPSC-derived MNs, dipeptide repeat proteins (DPRs)—encoded by the C9ORF72 hexanucleotide repeat expansions—preferentially bind to mitochondrial ribosomal proteins (Lopez-Gonzalez et al., 2016) thereby compromising mitochondrial function; it is likely DPRs have similar effects in astrocytes. Mutations in VCP (both ALS- and non-ALS-related) alter mitochondrial function in hiPSC-derived cortical neurons (Ludtmann et al., 2017) and increase ROS production in hiPSC-derived glial progenitor cells (Hall et al., 2017). Finally, the dysregulation of several mitochondrial genes in hiPSC-derived MNs from sALS patients suggests that mitochondrial dysfunction is a feature observed across ALS patient subgroups, though additional work must be done in order to establish whether this is also the case in astrocytes (Alves et al., 2015).

The pathological accumulation of ROS may not be solely due to mitochondrial dysfunction. For example, ROS produced by NADPH oxidase 2 (NOX2) enzymes is heightened in ALS patients, as NOX2 expression is progressively upregulated over time in SOD1(G93A) mice (Marden et al., 2007; Wu et al., 2006). Heterozygous deletion of NOX2 in SOD1(G93A) mice slows the progression of ALS-like phenotypes and increases animal survival, indicating that NOX2 suppression is a potential therapeutic target to slow disease progression (Reviewed by Bedard and Krause, 2007; Marden et al., 2007; Wu et al., 2006). The upregulation of NOX2 in SOD1 ALS mouse and primary astrocyte models (Marchetto et al., 2008; Wu et al., 2006) induces astrocyte reactivity and selective MN toxicity (Marchetto et al., 2008). Moreover, mSOD1 can also promote NOX2 activity by inhibiting Rac1 GTPase activity (Harraz et al., 2008). Interestingly, ALS2, which has been associated with juvenile ALS, is also a potent activator of RAC1 (Topp et al., 2004; Yang et al., 2001), although the effect of mutant ALS2 in astrocytes has not been examined.

These findings suggest that heightened ROS production is genetically encoded, and may even precede neurodegeneration (Park et al., 2009). Unfortunately, many of these genes, with the exception of SOD1 and VCP, have not yet been examined for their specific impact on astrocyte ROS production. This is especially crucial as mitochondrial proteins may display cell-type-specific heterogeneity in their contribution to ROS-related toxicity (Liu et al., 2004). Given that the aforementioned genes are relatively highly expressed in astrocytes, hiPSC models may enhance our understanding of how alterations to the functional development of astrocytes lays the foundation for ROS pathology later in life.

Excitotoxicity in ALS

Excitotoxic death is typically driven by the prolonged exposure of neurons to high levels of glutamate. The effects of excitotoxicity are mediated through Ca2+, which can be cytotoxic in large quantities. Accordingly, blocking Ca2+ signaling protects motor neurons (Beers et al., 2001; Van Den Bosch et al., 2002). Because of its ability to promote cell death in highly localized regions, the impact of excitotoxic mechanisms on ALS pathology is of particular interest, given that cell death is largely restricted to a particular neuronal population.

Astrocyte dysfunction contributes to excitotoxic conditions

Astrocytes are involved in the homeostatic-maintenance of extracellular ion and glutamate concentrations, which are crucial for ensuring normal neuronal excitability (Dehnes et al., 1998; Karwoski et al., 1989). In ALS pathology, this key function of astrocytes may be disrupted (Pamela J. Shaw et al., 1995; Spreux-Varoquaux et al., 2002). Taken together with the fact that the only medication to date that is prescribed for the treatment of ALS is Riluzole—an inhibitor of presynaptic glutamate release—this highlights that excitotoxic cell death is involved in ALS-related neurodegeneration. Of course, given that Riluzole is limited in its capacity to delay disease progression, this is likely not the only factor involved in MN death (Bensimon et al., 1996).

Astrocytes may promote excitotoxic conditions through decreased capacity for glutamate reuptake (Guo et al., 2003; Howland et al., 2002; Rothstein et al., 1995, 1992) or by glutamate release (Bezzi et al., 1998; Porter and McCarthy, 1996). Decreased expression of the astrocyte glutamate transporter EAAT2 has been observed in the brains and spinal cords of ALS patients (Fray et al., 1998; Rothstein et al., 1995, 1992). Gliotransmission, the astrocytic release of glutamate, may be altered in ALS patients. In the healthy brain, gliotransmission is thought to bolster synaptic plasticity (Reviewed by Schipke and Kettenmann, 2004), although the extent to which gliotransmission occurs in humans remains in question (Sloan and Barres, 2014). In vitro animal models suggest several potential mechanisms of glutamate release by astrocytes (Bezzi et al., 2004, 2001; Duan et al., 2003; Kimelberg et al., 1990; Takano et al., 2005; Ye et al., 2003) (Figure 2). For example, the activation of astrocytic CXCR4 chemokine receptors or purinergic P2Y1 receptors by pro-inflammatory mediators— including those produced by astrocytes, such as TNFα—induces Ca2+-mediated glutamate exocytosis in astrocytes (Bezzi et al., 2001, 1998; Domercq et al., 2006; Santello et al., 2011). In accordance with this mechanism, the overproduction of pro-inflammatory factors, as is seen in pathological neuroinflammation, is accompanied by an overall shift in synaptic glutamate regulation (Bezzi et al., 2001, 1998). Astrocytes can also release glutamate—as well as ATP—through connexin-43 (Cx-43) hemichannels (Ye et al., 2003). In addition to the obvious implications of this on excitotoxicity, glutamate and ATP released from astrocytes function together to increase permeability of neuronal pannexin-1 hemichannels for ATP, subsequently inducing apoptotic cell death in MNs (Almad et al., 2016; Orellana Juan A. et al., 2011). Importantly, ALS patients show heightened levels of Cx-43 in CNS tissue, and these results have been recapitulated in mice harboring the SOD1(G93A) mutation (Almad et al., 2016).

MNs may be more susceptible to excitotoxicity than other neurons even in healthy controls. There are a number of reasons for this, including a relatively low expression of GluR2-containing AMPA/kainate channels, rendering MNs more permeable to Ca2+. MNs also express sparse levels of intracellular Ca2+-buffering proteins (Ince et al., 1993; Takuma et al., 1999; Van Damme et al., 2002; Vandenberghe et al., 2000). Under non-pathological conditions, astrocytes modulate MN excitability, in part by regulating the number of GluR2-containing AMPA receptors expressed by MNs (Van Damme et al., 2007). Yet ALS patients display enhanced MN hyper-excitability, even in the pre-symptomatic and early stages of ALS pathology, suggesting a loss of such astrocytic modulatory input (Van Damme et al., 2007; Vucic et al., 2008; Vucic and Kiernan, 2006). Moreover, the heightened release of D-serine—an endogenous co-activator of NMDA receptors that is elevated in familial and sporadic ALS patients—by diseased astrocytes further enhances Ca2+ influx into MNs (Mitchell et al., 2010; Sasabe et al., 2012, 2007). Finally, NOX2-derived ROS (e.g. from astrocytes) can increase the conductance of L-type voltage gated calcium channels and contribute to MN hyperexcitability (Wang et al., 2004; Zimmerman et al., 2005). Thus, it appears that astrocytes are not only pertinent to the generation of excitotoxic conditions, but also are associated with increased MN sensitivity upon exposure to these conditions.

Hyperexcitability of MNs has been recapitulated in hiPSC-derived MNs from patients with SOD1, FUS, and C9ORF72 mutations (Wainger et al., 2014, Selvaraj et al., 2018). However, whether astrocytes contribute to hyperexcitability of hiPSC-derived MNs in vitro remains relatively unexplored. Systematic analysis of MN excitability when co-cultured with hiPSC-derived astrocytes from various ALS patient backgrounds could elucidate which mutations impact MN excitability through a non-cell autonomous mechanism.

Protein aggregation in ALS

The presence of protein aggregates (also referred to as inclusions) is a well-documented pathological feature of ALS and other neurodegenerative diseases (Durham et al., 1997; Jang et al., 2017; Johnson et al., 2009; Jucker and Walker, 2013; Mori et al., 2013; Schwartz et al., 2014). Indeed, up to 95% of all ALS patients have ubiquitinated inclusions that contain TDP-43 in the CNS (Neumann et al., 2006); protein inclusions are observed in both neurons and glia in human postmortem tissue (Forsberg et al., 2011; Mackenzie et al., 2007; Nishihira et al., 2008). TDP-43 is a ubiquitously expressed heterogeneous nuclear ribonucleoprotein (hnRNP) important for RNA metabolism. Mislocalization of TDP-43 from the nucleus to the cytoplasm and its subsequent sequestration within aggregates has been proposed as loss-of-function mechanism that disrupts its function in RNA metabolism (Scotter et al., 2015). Inclusions may also contain other RBPs and related hnRNPs (Conlon et al., 2018). Innovative computational approaches have proposed several new RBPs that could be mislocalized in ALS (Bakkar et al., 2018), all of which could be tested in hiPSC models.

The genesis and spread of inclusions remain an intensely studied topic and is beyond the scope of this review. However, recent studies suggest several possibilities. First, protein aggregation may be mediated by the interaction of so-called low complexity domains (LCDs) involved in liquid-liquid phase transitions necessary for membrane-less organelle formation (reviewed by Taylor et al., 2016). P bodies, stress granules, and RNA transport granules are all examples of membrane-less organelle structures that assemble and disassemble in response to stimuli. These highly dynamic structures are critical components of normal RNA metabolism. LCDs are common in RNA binding proteins (e.g. FUS, TDP-43, hnRNPs), which may render them more susceptible to persistent and aberrant aggregation (Purice and Taylor, 2018). Second, ALS mutations identified in genes involved in autophagy (Table 1) indicate that dysregulation of protein homeostasis contributes to aggregation. Autophagy, a major pathway for breaking down organelles, is important for the normal recycling of stress granules. Lastly, protein aggregates may be nucleated by DNA damage. DNA breaks have been linked to the mislocalization and aggregation of FUS in hiPSC-derived motor neurons (Naumann et al., 2018). Persistent protein aggregates may be toxic to cells; several mechanisms have been proposed (Gao et al., 2018; Scotter et al., 2015) that result in alterations to key components of cellular homeostasis, gene expression, and signal transduction (reviewed by Kim and Taylor, 2017).

Aggregation in hiPSC-derived motor neurons and astrocytes

ALS-related protein aggregation phenotypes observed in mice and human postmortem tissue have been difficult to recapitulate in hiPSC-derived MNs (reviewed in Lee and Huang, 2017). Even when using patient lines harboring mutations in TDP-43, TDP-43 inclusions in hiPSC-derived MNs are inconsistent across studies (Egawa et al., 2012; Fujimori et al., 2018; Seminary et al., 2018). However, inclusions have been observed in C9ORF72 (Donnelly et al., 2013; Sareen et al., 2013), SOD1 (Chen et al., 2014), and FUS (Purice and Taylor, 2018) patient lines. Repeat-associated non-ATG (RAN) translation of RNA from the C9ORF72 repeat produces several dipeptide repeat proteins (DPRs) that are aggregation-prone and proposed to interfere with nucleocytoplasmic transport (Reviewed by Yuva-Aydemir et al., 2018) and heterochromatin structure (Zhang et al., 2019). Consistent aggregation phenotypes, particularly in cells derived from sALS patients, may require more ‘aged’ cells (discussed in Future Directions) or other manipulation to mimic age-related phenotypes.

Few studies have examined whether aggregation occurs in glia derived from ALS hiPSCs. Cytoplasmic mislocalization of M337V TDP-43 has been observed in patient hiPSC-astrocytes as well as control astrocytes transduced with other mutant TDP-43 (Serio et al., 2013). These astrocytes showed decreased viability, but unlike SOD1 astrocytes, they did not impact MN survival in co-culture experiments. Given the divergent function of MNs and astrocytes, the repertoire of RNAs and LCD-containing RNA binding proteins found within membrane-less organelles in astrocytes may be different than in MNs. Unlike motor neurons that ultimately die during the course of disease, astrocytes proliferate and contribute to an inflammatory response. Thus, hiPSC-astrocytes could be used to model perturbations in astrocyte function as a result of intrinsic protein aggregation. For example, protein aggregate formation might induce a more toxic astrocyte state rather than cell death.

Murine models of ALS indicate astrocyte-dependent, non-cell autonomous impacts on MN degeneration in vivo (Clement et al., 2003) and in vitro (Phatnani et al., 2013). hiPSCs provide an ideal platform to investigate non-cell autonomous effects in vitro and have largely supported observations from murine models (Haidet-Phillips et al., 2011; Marchetto et al., 2008; Meyer et al., 2014). How inclusions observed in astrocytes exert effects on MNs is unclear. Endocytosis of SOD1 aggregates and subsequent toxicity to hiPSC-MNs supports a prion-like spread of disease (Benkler et al., 2018). Whether these aggregates originate and can be secreted by glia or MNs remains an open question. However, micro-RNAs secreted by C9ORF72 hiPSC-derived astrocytes that reduce MN viability in vitro have been observed (Varcianna et al., 2019). C9ORF72 hiPSC-MNs display cell-to-cell spreading of aggregates (Westergard et al., 2016). Additionally, SOD1 aggregates released by primary mouse astrocytes harboring the SOD1(G93A) mutation are taken up by MNs in co-culture and induce MN cell death, indicating a potential mechanism by which protein aggregation in astrocytes can promote neurodegeneration (Basso et al., 2013). TDP-43 aggregates are also transferred from cell-to-cell via exosomes, though it is unclear if TDP-43 containing exosomes could be astrocytic in origin (Nonaka et al., 2013). hiPSC-derived three-dimensional culture systems, which contain networks of neurons and glia, recapitulate aspects of Alzheimer’s disease pathology, including phosphorylated tau aggregates (Choi et al., 2014), and so may provide a system for investigating TDP-43 proteinopathy and the intercellular spread of inclusions.

Future Directions and Concluding Remarks

Due to their ability to recapitulate the complete genetic profile of the original donor fibroblast, hiPSC models allow for a new perspective in examining complex genetic diseases such as ALS. Unfortunately, despite the fact that a large portion of ALS-associated genes are highly expressed in astrocytes, only a handful of studies have applied this technique to study astrocyte involvement in ALS pathology. This is in contrast to hiPSC-derived MNs, which have been extensively modeled since the advent of hiPSCs. Moreover, of the studies that have been conducted, very few have examined those less penetrant genes that, unlike SOD1 and C9ORF72, are understudied in animal models. In particular, genes that have been associated with ALS pathology and are relatively highly expressed in fetal astrocytes are particularly well-suited for hiPSC modeling, due to the fetal expression profile of hiPSC-derived cells.

Still, the immature expression profile of hiPSC-derived cells brings into question the utility of hiPSC-derived astrocytes to model age-related disease pathology. One possible solution may be the development of protocols for the direct induction of specific cell types from patient fibroblasts, which would bypass stem-cell reprogramming and the resultant loss of age-related epigenetic markers. Therefore, the further development of artificial-aging and direct-induction protocols would enhance the utility of hiPSC-based systems to study disease pathology (Reviewed by Cornacchia and Studer, 2017). As the current understanding of astrocyte heterogeneity and patterning continues to improve (Reviewed by Farmer and Murai, 2017), the development of novel hiPSC differentiation techniques to better specify astrocyte subpopulations will be an important step towards improving hiPSC modeling of ALS. Although astrocytes have the capacity to alternate between protective and toxic phenotypes in vivo, this feature has yet to be accounted for in either hiPSC models. Future iPSC-derived astrocyte models, then, should profile astrocytes as they mature in co-culture, with a particular focus on understanding the relative abundance of protective versus toxic astrocytes present in culture. Conversely, controlling for this phenotype, perhaps by artificially driving the NF-kB or STAT3 pathways in patient-derived astrocytes, may enhance the reproducibility of in vitro ALS astrocyte models.

A better understanding of the relationship between astrocytes and ALS pathology may shed some light on other age-related neurodegenerative diseases. Patients with Alzheimer’s disease and Parkinson’s disease also frequently present with heightened levels of neuro-inflammation (Heneka et al., 2015; Wang et al., 2015). Even more specifically, certain disease-related phenotypes, such as the upregulation of Cx-43, have been noted in these diseases as well, suggesting that potential pathological spread of disease via astrocyte gap junctions may be shared (Kawasaki et al., 2009, p. 43; Yi et al., 2016). Overall, consideration of astrocyte-mediated roles in neurodegeneration could increase the number of potential therapeutic targets for these diseases.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Abbott NJ, 2000. Inflammatory Mediators and Modulation of Blood–Brain Barrier Permeability. Cell Mol Neurobiol 20, 131–147. 10.1023/A:1007074420772 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Ajroud-Driss S, Fecto F, Ajroud K, Lalani I, Calvo SE, Mootha VK, Deng H-X, Siddique N, Tahmoush AJ, Heiman-Patterson TD, Siddique T, 2015. Mutation in the novel nuclear-encoded mitochondrial protein CHCHD10 in a family with autosomal dominant mitochondrial myopathy. Neurogenetics 16, 1–9. 10.1007/s10048-014-0421-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Alami NH, Smith RB, Carrasco MA, Williams LA, Winborn CS, Han SSW, Kiskinis E, Winborn B, Freibaum BD, Kanagaraj A, Clare AJ, Badders NM, Bilican B, Chaum E, Chandran S, Shaw CE, Eggan KC, Maniatis T, Taylor JP, 2014. Axonal Transport of TDP-43 mRNA Granules Is Impaired by ALS-Causing Mutations. Neuron 81, 536–543. 10.1016/j.neuron.2013.12.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Alexianu ME, Kozovska M, Appel SH, 2001. Immune reactivity in a mouse model of familial ALS correlates with disease progression. Neurology 57, 1282–1289. [DOI] [PubMed] [Google Scholar]
  5. Allen SP, Hall B, Castelli LM, Francis L, Woof R, Siskos AP, Kouloura E, Gray E, Thompson AG, Talbot K, Higginbottom A, Myszczynska M, Allen CF, Stopford MJ, Hemingway J, Bauer CS, Webster CP, De Vos KJ, Turner MR, Keun HC, Hautbergue GM, Ferraiuolo L, Shaw PJ, 2019. Astrocyte adenosine deaminase loss increases motor neuron toxicity in amyotrophic lateral sclerosis. Brain 142, 586–605. 10.1093/brain/awy353 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Almad AA, Doreswamy A, Gross SK, Richard J-P, Huo Y, Haughey N, Maragakis NJ, 2016. Connexin 43 in astrocytes contributes to motor neuron toxicity in amyotrophic lateral sclerosis. Glia 64, 1154–1169. 10.1002/glia.22989 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Alves CJ, Dariolli R, Jorge FM, de H, Monteiro MR, Maximino JR, Martins RS, Strauss BE, Krieger JE, Callegaro D, Chadi G, 2015. Gene expression profiling for human iPS-derived motor neurons from sporadic ALS patients reveals a strong association between mitochondrial functions and neurodegeneration. Front. Cell. Neurosci 9 10.3389/fncel.2015.00289 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Arthur KC, Calvo A, Price TR, Geiger JT, Chiò A, Traynor BJ, 2016. Projected increase in amyotrophic lateral sclerosis from 2015 to 2040. Nat Commun 7 10.1038/ncomms12408 [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Atanasio A, Decman V, White D, Ramos M, Ikiz B, Lee HC, Siao CJ, Brydges S, LaRosa E, Bai Y, Fury W, Burfeind P, Zamfirova R, Warshaw G, 2016. C9orf72 ablation causes immune dysregulation characterized by leukocyte expansion, autoantibody production, and glomerulonephropathy in mice. - PubMed - NCBI. Sci Rep 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Atkins CM, Chen SJ, Klann E, Sweatt JD, 1997. Increased phosphorylation of myelin basic protein during hippocampal long-term potentiation. J. Neurochem 68, 1960–1967. [DOI] [PubMed] [Google Scholar]
  11. Atkins CM, Sweatt JD, 1999. Reactive oxygen species mediate activity-dependent neuron-glia signaling in output fibers of the hippocampus. J. Neurosci 19, 7241–7248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Bakkar N, Kovalik T, Lorenzini I, Spangler S, Lacoste A, Sponaugle K, Ferrante P, Argentinis E, Sattler R, Bowser R, 2018. Artificial intelligence in neurodegenerative disease research: use of IBM Watson to identify additional RNA-binding proteins altered in amyotrophic lateral sclerosis. Acta Neuropathol 135, 227–247. 10.1007/s00401-017-1785-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Bannwarth S, Ait-El-Mkadem S, Chaussenot A, Genin EC, Lacas-Gervais S, Fragaki K, Berg-Alonso L, Kageyama Y, Serre V, Moore DG, Verschueren A, Rouzier C, Le Ber I, Augé G, Cochaud C, Lespinasse F, N’Guyen K, de Septenville A, Brice A, Yu-Wai-Man P, Sesaki H, Pouget J, Paquis-Flucklinger V, 2014. A mitochondrial origin for frontotemporal dementia and amyotrophic lateral sclerosis through CHCHD10 involvement. Brain 137, 2329–2345. 10.1093/brain/awu138 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Barmada SJ, Serio A, Arjun A, Bilican B, Daub A, Ando DM, Tsvetkov A, Pleiss M, Li X, Peisach D, Shaw C, Chandran S, Finkbeiner S, 2014. Autophagy induction enhances TDP43 turnover and survival in neuronal ALS models. Nature Chemical Biology; Cambridge: 10, 677–85. http://dx.doi.org.ezproxy.cul.columbia.edu/10.1038/nchembio.1563 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Barres BA, 2008. The Mystery and Magic of Glia: A Perspective on Their Roles in Health and Disease. Neuron 60, 430–440. 10.1016/j.neuron.2008.10.013 [DOI] [PubMed] [Google Scholar]
  16. Basso M, Pozzi S, Tortarolo M, Fiordaliso F, Bisighini C, Pasetto L, Spaltro G, Lidonnici D, Gensano F, Battaglia E, Bendotti C, Bonetto V, 2013. Mutant copper-zinc superoxide dismutase (SOD1) induces protein secretion pathway alterations and exosome release in astrocytes: implications for disease spreading and motor neuron pathology in amyotrophic lateral sclerosis. J. Biol. Chem jbc.M112.425066. 10.1074/jbc.M112.425066 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bedard K, Krause K-H, 2007. The NOX Family of ROS-Generating NADPH Oxidases: Physiology and Pathophysiology. Physiological Reviews 87, 245–313. 10.1152/physrev.00044.2005 [DOI] [PubMed] [Google Scholar]
  18. Beers DR, Ho B-K, Siklós L, Alexianu ME, Mosier DR, Mohamed AH, Otsuka Y, Kozovska ME, McAlhany RE, Smith RG, Appel SH, 2001. Parvalbumin overexpression alters immune-mediated increases in intracellular calcium, and delays disease onset in a transgenic model of familial amyotrophic lateral sclerosis. Journal of Neurochemistry 79, 499–509. 10.1046/j.1471-4159.2001.00582.x [DOI] [PubMed] [Google Scholar]
  19. Benkler C, O’Neil AL, Slepian S, Qian F, Weinreb PH, Rubin LL, 2018. Aggregated SOD1 causes selective death of cultured human motor neurons. Scientific Reports 8, 16393 10.1038/s41598-018-34759-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Bensimon G, Leigh PN, Guillet P, Lacomblez L, 1996. Dose-ranging study of riluzole in amyotrophic lateral sclerosis. The Lancet 347, 1425–1431. 10.1016/S0140-6736(96)91680-3 [DOI] [PubMed] [Google Scholar]
  21. Bezzi P, Carmignoto G, Pasti L, Vesce S, Rossi D, Rizzini BL, Pozzan T, Volterra A, 1998. Prostaglandins stimulate calcium-dependent glutamate release in astrocytes. Nature 391, 281–285. 10.1038/34651 [DOI] [PubMed] [Google Scholar]
  22. Bezzi P, Domercq M, Brambilla L, Galli R, Schols D, De Clercq E, Vescovi A, Bagetta G, Kollias G, Meldolesi J, Volterra A, 2001. CXCR4-activated astrocyte glutamate release via TNFα: amplification by microglia triggers neurotoxicity. Nature Neuroscience 4, 702–710. 10.1038/89490 [DOI] [PubMed] [Google Scholar]
  23. Bezzi P, Gundersen V, Galbete J, Seifert G, Steinher C, Pilati E, Volterra A, 2004. Astrocytes contain a vesicular compartment that is competent for regulated exocytosis of glutamate. Nature Neuroscience; New York: 7, 613–20. http://dx.doi.org.ezproxy.cul.columbia.edu/10.1038/nn1246 [DOI] [PubMed] [Google Scholar]
  24. Bhinge A, Namboori SC, Zhang X, VanDongen AMJ, Stanton LW, 2017. Genetic Correction of SOD1 Mutant iPSCs Reveals ERK and JNK Activated AP1 as a Driver of Neurodegeneration in Amyotrophic Lateral Sclerosis. Stem Cell Reports 8, 856–869. 10.1016/j.stemcr.2017.02.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Bilican B, Serio A, Barmada SJ, Nishimura AL, Sullivan GJ, Carrasco M, Phatnani HP, Puddifoot CA, Story D, Fletcher J, Park I-H, Friedman BA, Daley GQ, Wyllie DJA, Hardingham GE, Wilmut I, Finkbeiner S, Maniatis T, Shaw CE, Chandran S, 2012. Mutant induced pluripotent stem cell lines recapitulate aspects of TDP-43 proteinopathies and reveal cell-specific vulnerability. PNAS 109, 5803–5808. 10.1073/pnas.1202922109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Boillée S, Velde CV, Cleveland DW, 2006. ALS: A Disease of Motor Neurons and Their Nonneuronal Neighbors. Neuron 52, 39–59. 10.1016/j.neuron.2006.09.018 [DOI] [PubMed] [Google Scholar]
  27. Bosco DA, Morfini G, Karabacak NM, Song Y, Gros-louis F, Pasinelli P, Goolsby H, Fontaine BA, Lemay N, Mckenna-yasek D, Frosch MP, Agar JN, Julien J, Brady ST, Brown RH, 2010. Wild-type and mutant SOD1 share an aberrant conformation and a common pathogenic pathway in ALS. Nature Neuroscience; New York: 13, 1396–403. http://dx.doi.org.ezproxy.cul.columbia.edu/10.1038/nn.2660 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Brambilla R, Bracchi-Ricard V, Hu W-H, Frydel B, Bramwell A, Karmally S, Green EJ, Bethea JR, 2005. Inhibition of astroglial nuclear factor κB reduces inflammation and improves functional recovery after spinal cord injury. Journal of Experimental Medicine 202, 145–156. 10.1084/jem.20041918 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Brambilla R, Dvoriantchikova G, Barakat D, Ivanov D, Bethea JR, Shestopalov VI, 2012. Transgenic inhibition of astroglial NF-κB protects from optic nerve damage and retinal ganglion cell loss in experimental optic neuritis. Journal of Neuroinflammation 9, 213 10.1186/1742-2094-9-213 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Brambilla R, Persaud T, Hu X, Karmally S, Shestopalov VI, Dvoriantchikova G, Ivanov D, Nathanson L, Barnum SR, Bethea JR, 2009. Transgenic Inhibition of Astroglial NF-κB Improves Functional Outcome in Experimental Autoimmune Encephalomyelitis by Suppressing Chronic Central Nervous System Inflammation. The Journal of Immunology 182, 2628–2640. 10.4049/jimmunol.0802954 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Braun RJ, Sommer C, Carmona-Gutierrez D, Khoury CM, Ring J, Büttner S, Madeo F, 2011. Neurotoxic 43-kDa TAR DNA-binding Protein (TDP-43) Triggers Mitochondrion-dependent Programmed Cell Death in Yeast. J. Biol. Chem 286, 19958–19972. 10.1074/jbc.M110.194852 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Bruijn LI, Becher MW, Lee MK, Anderson KL, Jenkins NA, Copeland NG, Sisodia SS, Rothstein JD, Borchelt DR, Price DL, Cleveland DW, 1997. ALS-Linked SOD1 Mutant G85R Mediates Damage to Astrocytes and Promotes Rapidly Progressive Disease with SOD1-Containing Inclusions. Neuron 18, 327–338. 10.1016/S0896-6273(00)80272-X [DOI] [PubMed] [Google Scholar]
  33. Cadenas E, Boveris A, Ragan CI, Stoppani AOM, 1977. Production of superoxide radicals and hydrogen peroxide by NADH-ubiquinone reductase and ubiquinol-cytochrome c reductase from beef-heart mitochondria. Archives of Biochemistry and Biophysics 180, 248–257. 10.1016/0003-9861(77)90035-2 [DOI] [PubMed] [Google Scholar]
  34. Caiazzo M, Giannelli S, Valente P, Lignani G, Carissimo A, Sessa A, Colasante G, Bartolomeo R, Massimino L, Ferroni S, Settembre C, Benfenati F, Broccoli V, 2014. Direct Conversion of Fibroblasts into Functional Astrocytes by Defined Transcription Factors. Stem Cell Reports 4, 25–36. 10.1016/j.stemcr.2014.12.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Canals I, Ginisty A, Quist E, Timmerman R, Fritze J, Miskinyte G, Monni E, Hansen MG, Hidalgo I, Bryder D, Bengzon J, Ahlenius H, 2018. Rapid and Efficient Induction of Functional Astrocytes from Human Pluripotent Stem Cells [DOI] [PubMed] [Google Scholar]
  36. Cassina P, Cassina A, Pehar M, Castellanos R, Gandelman M, León A. de, Robinson KM, Mason RP, Beckman JS, Barbeito L, Radi R, 2008. Mitochondrial Dysfunction in SOD1G93A-Bearing Astrocytes Promotes Motor Neuron Degeneration: Prevention by Mitochondrial-Targeted Antioxidants. J. Neurosci 28, 4115–4122. 10.1523/JNEUROSCI.5308-07.2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Chao CC, Hu S, Sheng WS, Bu D, Bukrinsky MI, Peterson PK, 1996. Cytokine-stimulated astrocytes damage human neurons via a nitric oxide mechanism. Glia 16, 276–284. [DOI] [PubMed] [Google Scholar]
  38. Che X-Q, Zhao Q-H, Huang Y, Li X, Ren R-J, Chen S-D, Wang G, Guo Q-H, 2017. Genetic Features of MAPT, GRN, C9orf72 and CHCHD10 Gene Mutations in Chinese Patients with Frontotemporal Dementia. Curr Alzheimer Res 14, 1102–1108. 10.2174/1567205014666170426105713 [DOI] [PubMed] [Google Scholar]
  39. Chen H, Kankel MW, Su SC, Han SWS, Ofengeim D, 2018. Exploring the genetics and non-cell autonomous mechanisms underlying ALS/FTLD. Cell Death & Differentiation 1 10.1038/s41418-018-0060-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Chen H, Qian K, Du Z, Cao J, Petersen A, Liu H, Blackbourn LW, Huang C-L, Errigo A, Yin Y, Lu J, Ayala M, Zhang S-C, 2014. Modeling ALS with iPSCs Reveals that Mutant SOD1 Misregulates Neurofilament Balance in Motor Neurons. Cell Stem Cell 14, 796–809. 10.1016/j.stem.2014.02.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Chen Y, Chen C-F, Riley DJ, Chen P-L, 2011. Nek1 kinase functions in DNA damage response and checkpoint control through a pathway independent of ATM and ATR. Cell Cycle 10, 655–663. 10.4161/cc.10.4.14814 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Cherry JD, Olschowka JA, O’Banion MK, 2014. Neuroinflammation and M2 microglia: the good, the bad, and the inflamed. Journal of Neuroinflammation 11, 98 10.1186/1742-2094-11-98 [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Ching S, Zhang H, Belevych N, He L, Lai W, Pu X, Jaeger LB, Chen Q, Quan N, 2007. Endothelial-Specific Knockdown of Interleukin-1 (IL-1) Type 1 Receptor Differentially Alters CNS Responses to IL-1 Depending on Its Route of Administration. J. Neurosci 27, 10476–10486. 10.1523/JNEUROSCI.3357-07.2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Chiò A, Borghero G, Restagno G, Mora G, Drepper C, Traynor BJ, Sendtner M, Brunetti M, Ossola I, Calvo A, Pugliatti M, Sotgiu MA, Murru MR, Marrosu MG, Marrosu F, Marinou K, Mandrioli J, Sola P, Caponnetto C, Mancardi G, Mandich P, La Bella V, Spataro R, Conte A, Monsurrò MR, Tedeschi G, Pisano F, Bartolomei I, Salvi F, Lauria Pinter G, Simone I, Logroscino G, Gambardella A, Quattrone A, Lunetta C, Volanti P, Zollino M, Penco S, Battistini S, Renton AE, Majounie E, Abramzon Y, Conforti FL, Giannini F, Corbo M, Sabatelli M, 2012. Clinical characteristics of patients with familial amyotrophic lateral sclerosis carrying the pathogenic GGGGCC hexanucleotide repeat expansion of C9ORF72. Brain 135, 784–793. 10.1093/brain/awr366 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Choi SH, Kim YH, Hebisch M, Sliwinski C, Lee S, D’Avanzo C, Chen H, Hooli B, Asselin C, Muffat J, Klee JB, Zhang C, Wainger BJ, Peitz M, Kovacs DM, Woolf CJ, Wagner SL, Tanzi RE, Kim DY, 2014. A three-dimensional human neural cell culture model of Alzheimer’s disease. Nature 515, 274–278. 10.1038/nature13800 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Cirulli ET, Lasseigne BN, Petrovski S, Sapp PC, Dion PA, Leblond CS, Couthouis J, Lu Y-F, Wang Q, Krueger BJ, Ren Z, Keebler J, Han Y, Levy SE, Boone BE, Wimbish JR, Waite LL, Jones AL, Carulli JP, Day-Williams AG, Staropoli JF, Xin WW, Chesi A, Raphael AR, McKenna-Yasek D, Cady J, Jong J.M.B.V. de, Kenna KP, Smith BN, Topp S, Miller J, Gkazi A, Consortium, F.S., Al-Chalabi A, van den Berg LH, Veldink J, Silani V, Ticozzi N, Shaw CE, Baloh RH, Appel S, Simpson E, Lagier-Tourenne C, Pulst SM, Gibson S, Trojanowski JQ, Elman L, McCluskey L, Grossman M, Shneider NA, Chung WK, Ravits JM, Glass JD, Sims KB, Deerlin VMV, Maniatis T, Hayes SD, Ordureau A, Swarup S, Landers J, Baas F, Allen AS, Bedlack RS, Harper JW, Gitler AD, Rouleau GA, Brown R, Harms MB, Cooper GM, Harris T, Myers RM, Goldstein DB, 2015. Exome sequencing in amyotrophic lateral sclerosis identifies risk genes and pathways. Science 347, 1436–1441. 10.1126/science.aaa3650 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Clarke LE, Barres BA, 2013. Emerging roles of astrocytes in neural circuit development. Nature Reviews Neuroscience 14, 311–321. 10.1038/nrn3484 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Clement AM, Nguyen MD, Roberts EA, Garcia ML, Boillée S, Rule M, McMahon AP, Doucette W, Siwek D, Ferrante RJ, Brown RH, Julien J-P, Goldstein LSB, Cleveland DW, 2003. Wild-Type Nonneuronal Cells Extend Survival of SOD1 Mutant Motor Neurons in ALS Mice. Science 302, 113–117. 10.1126/science.1086071 [DOI] [PubMed] [Google Scholar]
  49. Cohen TJ, Hwang AW, Unger T, Trojanowski JQ, Lee VMY, 2012. Redox signalling directly regulates TDP‐43 via cysteine oxidation and disulphide cross‐linking. The EMBO Journal 31, 1241–1252. 10.1038/emboj.2011.471 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Colombo E, Cordiglieri C, Melli G, Newcombe J, Krumbholz M, Parada LF, Medico E, Hohlfeld R, Meinl E, Farina C, 2012. Stimulation of the neurotrophin receptor TrkB on astrocytes drives nitric oxide production and neurodegeneration. Journal of Experimental Medicine 209, 521–535. 10.1084/jem.20110698 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Conlon EG, Fagegaltier D, Agius P, Davis-Porada J, Gregory J, Hubbard I, Kang K, Kim D, New York Genome Center ALS Consortium, Phatnani H, Kwan J, Sareen D, Broach JR, Simmons Z, Arcila-Londono X, Lee EB, Van Deerlin VM, Shneider NA, Fraenkel E, Ostrow LW, Baas F, Zaitlen N, Berry JD, Malaspina A, Fratta P, Cox GA, Thompson LM, Finkbeiner S, Dardiotis E, Miller TM, Chandran S, Pal S, Hornstein E, MacGowan DJ, Heiman-Patterson T, Hammell MG, Patsopoulos NA, Dubnau J, Nath A, Phatnani H, Shneider NA, Manley JL, 2018. Unexpected similarities between C9ORF72 and sporadic forms of ALS/FTD suggest a common disease mechanism. Elife 7 10.7554/eLife.37754 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Cooper-Knock J, Moll T, Ramesh T, Castelli L, Beer A, Robins H, Fox I, Niedermoser I, Van Damme P, Moisse M, Robberecht W, Hardiman O, Panades MP, Assialioui A, Mora JS, Basak AN, Morrison KE, Shaw CE, Al-Chalabi A, Landers JE, Wyles M, Heath PR, Higginbottom A, Walsh T, Kazoka M, McDermott CJ, Hautbergue GM, Kirby J, Shaw PJ, 2019. Mutations in the Glycosyltransferase Domain of GLT8D1 Are Associated with Familial Amyotrophic Lateral Sclerosis. Cell Rep 26, 2298–2306.e5. 10.1016/j.celrep.2019.02.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Coppedè F, Migliore L, 2015. DNA damage in neurodegenerative diseases. Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis, DNA damage in chronic diseases and aging 776, 84–97. 10.1016/j.mrfmmm.2014.11.010 [DOI] [PubMed] [Google Scholar]
  54. Cornacchia D, Studer L, 2017. Back and forth in time: Directing age in iPSC-derived lineages. Brain Research 1656, 14–26. 10.1016/j.brainres.2015.11.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Corral-Debrinski M, Horton T, Lott MT, Shoffner JM, Flint Beal M, Wallace DC, 1992. Mitochondrial DNA deletions in human brain: regional variability and increase with advanced age. Nature Genetics 2, 324–329. 10.1038/ng1292-324 [DOI] [PubMed] [Google Scholar]
  56. Dantzer R, O’Connor JC, Freund GG, Johnson RW, Kelley KW, 2008. From inflammation to sickness and depression: when the immune system subjugates the brain. Nature Reviews Neuroscience 9, 46–56. 10.1038/nrn2297 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Dawson VL, Dawson TM, Bartley DA, Uhl GR, Snyder SH, 1993. Mechanisms of nitric oxide-mediated neurotoxicity in primary brain cultures. J. Neurosci 13, 2651–2661. 10.1523/JNEUROSCI.13-06-02651.1993 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. De Vos KJ, Mórotz GM, Stoica R, Tudor EL, Lau K-F, Ackerley S, Warley A, Shaw CE, Miller CCJ, 2012. VAPB interacts with the mitochondrial protein PTPIP51 to regulate calcium homeostasis. Hum Mol Genet 21, 1299–1311. 10.1093/hmg/ddr559 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Dehnes Y, Chaudhry FA, Ullensvang K, Lehre KP, Storm-Mathisen J, Danbolt NC, 1998. The Glutamate Transporter EAAT4 in Rat Cerebellar Purkinje Cells: A Glutamate-Gated Chloride Channel Concentrated near the Synapse in Parts of the Dendritic Membrane Facing Astroglia. J. Neurosci 18, 3606–3619. 10.1523/JNEUROSCI.18-10-03606.1998 [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. DeJesus-Hernandez M, Mackenzie IR, Boeve BF, Boxer AL, Baker M, Rutherford NJ, Nicholson AM, Finch NA, Flynn H, Adamson J, Kouri N, Wojtas A, Sengdy P, Hsiung G-YR, Karydas A, Seeley WW, Josephs KA, Coppola G, Geschwind DH, Wszolek ZK, Feldman H, Knopman DS, Petersen RC, Miller BL, Dickson DW, Boylan KB, Graff-Radford NR, Rademakers R, 2011. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 72, 245–256. 10.1016/j.neuron.2011.09.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Di Giorgio FP, Carrasco MA, Siao MC, Maniatis T, Eggan K, 2007. Non–cell autonomous effect of glia on motor neurons in an embryonic stem cell–based ALS model. Nature Neuroscience 10, 608–614. 10.1038/nn1885 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Ditsworth D, Maldonado M, McAlonis-Downes M, Sun S, Seelman A, Drenner K, Arnold E, Ling S-C, Pizzo D, Ravits J, Cleveland DW, Da Cruz S, 2017. Mutant TDP-43 within motor neurons drives disease onset but not progression in amyotrophic lateral sclerosis. Acta Neuropathol 133, 907–922. 10.1007/s00401-017-1698-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Domercq M, Brambilla L, Pilati E, Marchaland J, Volterra A, Bezzi P, 2006. P2Y1 Receptor-evoked Glutamate Exocytosis from Astrocytes CONTROL BY TUMOR NECROSIS FACTOR-α AND PROSTAGLANDINS. J. Biol. Chem 281, 30684–30696. 10.1074/jbc.M606429200 [DOI] [PubMed] [Google Scholar]
  64. Donnelly CJ, Zhang P-W, Pham JT, Haeusler AR, Mistry NA, Vidensky S, Daley EL, Poth EM, Hoover B, Fines DM, Maragakis N, Tienari PJ, Petrucelli L, Traynor BJ, Wang J, Rigo F, Bennett CF, Blackshaw S, Sattler R, Rothstein JD, 2013. RNA Toxicity from the ALS/FTD C9ORF72 Expansion Is Mitigated by Antisense Intervention. Neuron 80, 415–428. 10.1016/j.neuron.2013.10.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Downen M, Amaral TD, Hua LL, Zhao M-L, Lee SC, 1999. Neuronal death in cytokine-activated primary human brain cell culture: role of tumor necrosis factor-α. Glia 28, 114–127. [DOI] [PubMed] [Google Scholar]
  66. Duan S, Anderson CM, Keung EC, Chen Yongmei, Chen Yiren, Swanson RA, 2003. P2X7 Receptor-Mediated Release of Excitatory Amino Acids from Astrocytes. J. Neurosci 23, 1320–1328. 10.1523/JNEUROSCI.23-04-01320.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Duan W, Li X, Shi J, Guo Y, Li Z, Li C, 2010. Mutant TAR DNA-binding protein-43 induces oxidative injury in motor neuron-like cell. Neuroscience 169, 1621–1629. 10.1016/j.neuroscience.2010.06.018 [DOI] [PubMed] [Google Scholar]
  68. Durham HD, Roy J, Dong L, Figlewicz DA, 1997. Aggregation of Mutant Cu/Zn Superoxide Dismutase Proteins in a Culture Model of ALS. J Neuropathol Exp Neurol 56, 523–530. 10.1097/00005072-199705000-00008 [DOI] [PubMed] [Google Scholar]
  69. Egashira Y, Hua Y, Keep RF, Iwama T, Xi G, 2016. Lipocalin 2 and Blood-Brain Barrier Disruption in White Matter after Experimental Subarachnoid Hemorrhage. Acta Neurochir. Suppl 121, 131–134. 10.1007/978-3-319-18497-5_23 [DOI] [PubMed] [Google Scholar]
  70. Egawa N, Kitaoka S, Tsukita K, Naitoh M, Takahashi K, Yamamoto T, Adachi F, Kondo T, Okita K, Asaka I, Aoi T, Watanabe A, Yamada Y, Morizane A, Takahashi J, Ayaki T, Ito H, Yoshikawa K, Yamawaki S, Suzuki S, Watanabe D, Hioki H, Kaneko T, Makioka K, Okamoto K, Takuma H, Tamaoka A, Hasegawa K, Nonaka T, Hasegawa M, Kawata A, Yoshida M, Nakahata T, Takahashi R, Marchetto MCN, Gage FH, Yamanaka S, Inoue H, 2012. Drug Screening for ALS Using Patient-Specific Induced Pluripotent Stem Cells. Science Translational Medicine 4, 145ra104–145ra104. 10.1126/scitranslmed.3004052 [DOI] [PubMed] [Google Scholar]
  71. Ellwardt E, Zipp F, 2014. Molecular mechanisms linking neuroinflammation and neurodegeneration in MS. Experimental Neurology, Special Issue: Progress in MS pathophysiology and treatment 262, 8–17. 10.1016/j.expneurol.2014.02.006 [DOI] [PubMed] [Google Scholar]
  72. Emdad L, D’Souza SL, Kothari HP, Qadeer ZA, Germano IM, 2011. Efficient Differentiation of Human Embryonic and Induced Pluripotent Stem Cells into Functional Astrocytes. Stem Cells and Development 21, 404–410. 10.1089/scd.2010.0560 [DOI] [PubMed] [Google Scholar]
  73. Endo F, Komine O, Fujimori-Tonou N, Katsuno M, Jin S, Watanabe S, Sobue G, Dezawa M, Wyss-Coray T, Yamanaka K, 2015. Astrocyte-Derived TGF-β1 Accelerates Disease Progression in ALS Mice by Interfering with the Neuroprotective Functions of Microglia and T Cells. Cell Reports 11, 592–604. 10.1016/j.celrep.2015.03.053 [DOI] [PubMed] [Google Scholar]
  74. van Es MA, Dahlberg C, Birve A, Veldink JH, van den Berg LH, Andersen PM, 2010. Large-scale SOD1 mutation screening provides evidence for genetic heterogeneity in amyotrophic lateral sclerosis. Journal of Neurology, Neurosurgery & Psychiatry 81, 562–566. 10.1136/jnnp.2009.181453 [DOI] [PubMed] [Google Scholar]
  75. Ezzi SA, Urushitani M, Julien J-P, 2007. Wild-type superoxide dismutase acquires binding and toxic properties of ALS-linked mutant forms through oxidation. Journal of Neurochemistry 102, 170–178. 10.1111/j.1471-4159.2007.04531.x [DOI] [PubMed] [Google Scholar]
  76. Farg MA, Soo KY, Warraich ST, Sundaramoorthy V, Blair IP, Atkin JD, 2013. Ataxin-2 interacts with FUS and intermediate-length polyglutamine expansions enhance FUS-related pathology in amyotrophic lateral sclerosis. Hum Mol Genet 22, 717–728. 10.1093/hmg/dds479 [DOI] [PubMed] [Google Scholar]
  77. Farmer WT, Murai K, 2017. Resolving Astrocyte Heterogeneity in the CNS. Front Cell Neurosci 11 10.3389/fncel.2017.00300 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Ferraiuolo L, Higginbottom A, Heath PR, Barber S, Greenald D, Kirby J, Shaw PJ, 2011. Dysregulation of astrocyte–motoneuron cross-talk in mutant superoxide dismutase 1-related amyotrophic lateral sclerosis. Brain 134, 2627–2641. 10.1093/brain/awr193 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Ferrante RJ, Browne SE, Shinobu LA, Bowling AC, Baik MJ, MacGarvey U, Kowall NW, Brown RH, Beal MF, 1997. Evidence of increased oxidative damage in both sporadic and familial amyotrophic lateral sclerosis. J. Neurochem 69, 2064–2074. [DOI] [PubMed] [Google Scholar]
  80. Forsberg K, Andersen PM, Marklund SL, Brännström T, 2011. Glial nuclear aggregates of superoxide dismutase-1 are regularly present in patients with amyotrophic lateral sclerosis. Acta Neuropathol 121, 623–634. 10.1007/s00401-011-0805-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Frank MG, Barrientos RM, Biedenkapp JC, Rudy JW, Watkins LR, Maier SF, 2006. mRNA up-regulation of MHC II and pivotal pro-inflammatory genes in normal brain aging. Neurobiol. Aging 27, 717–722. 10.1016/j.neurobiolaging.2005.03.013 [DOI] [PubMed] [Google Scholar]
  82. Fray AE, Ince PG, Banner SJ, Milton ID, Usher PA, Cookson MR, Shaw PJ, 1998. The expression of the glial glutamate transporter protein EAAT2 in motor neuron disease: an immunohistochemical study. European Journal of Neuroscience 10, 2481–2489. 10.1046/j.1460-9568.1998.00273.x [DOI] [PubMed] [Google Scholar]
  83. Fujimori K, Ishikawa M, Otomo A, Atsuta N, Nakamura R, Akiyama T, Hadano S, Aoki M, Saya H, Sobue G, Okano H, 2018. Modeling sporadic ALS in iPSC-derived motor neurons identifies a potential therapeutic agent. Nature Medicine 24, 1579 10.1038/s41591-018-0140-5 [DOI] [PubMed] [Google Scholar]
  84. Fukunaga K, Shinoda Y, Tagashira H, 2015. The role of SIGMAR1 gene mutation and mitochondrial dysfunction in amyotrophic lateral sclerosis. Journal of Pharmacological Sciences 127, 36–41. 10.1016/j.jphs.2014.12.012 [DOI] [PubMed] [Google Scholar]
  85. Gao J, Wang L, Huntley ML, Perry G, Wang X, 2018. Pathomechanisms of TDP-43 in neurodegeneration. Journal of Neurochemistry 146, 7–20. 10.1111/jnc.14327 [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Genin EC, Plutino M, Bannwarth S, Villa E, Cisneros‐Barroso E, Roy M, Ortega‐Vila B, Fragaki K, Lespinasse F, Pinero‐Martos E, Augé G, Moore D, Burté F, Lacas‐ Gervais S, Kageyama Y, Itoh K, Yu‐Wai‐Man P, Sesaki H, Ricci J-E, Vives‐Bauza C, Paquis‐Flucklinger V, 2016. CHCHD10 mutations promote loss of mitochondrial cristae junctions with impaired mitochondrial genome maintenance and inhibition of apoptosis. EMBO Molecular Medicine 8, 58–72. 10.15252/emmm.201505496 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Gomez-Deza J, Lee Y, Troakes C, Nolan M, Al-Sarraj S, Gallo J-M, Shaw CE, 2015. Dipeptide repeat protein inclusions are rare in the spinal cord and almost absent from motor neurons in C9ORF72 mutant amyotrophic lateral sclerosis and are unlikely to cause their degeneration. Acta Neuropathologica Communications 3, 38 10.1186/s40478-015-0218-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Gong YH, Parsadanian AS, Andreeva A, Snider WD, Elliott JL, 2000. Restricted expression of G86R Cu/Zn superoxide dismutase in astrocytes results in astrocytosis but does not cause motoneuron degeneration. J. Neurosci 20, 660–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Guareschi S, Cova E, Cereda C, Ceroni M, Donetti E, Bosco DA, Trotti D, Pasinelli P, 2012. An over-oxidized form of superoxide dismutase found in sporadic amyotrophic lateral sclerosis with bulbar onset shares a toxic mechanism with mutant SOD1. PNAS 109, 5074–5079. 10.1073/pnas.1115402109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Guerreiro R, Brás J, Hardy J, 2015. SnapShot: Genetics of ALS and FTD. Cell 160, 798.e1. 10.1016/j.cell.2015.01.052 [DOI] [PubMed] [Google Scholar]
  91. Guo H, Lai L, Butchbach MER, Stockinger MP, Shan X, Bishop GA, Lin CG, 2003. Increased expression of the glial glutamate transporter EAAT2 modulates excitotoxicity and delays the onset but not the outcome of ALS in mice. Hum Mol Genet 12, 2519–2532. 10.1093/hmg/ddg267 [DOI] [PubMed] [Google Scholar]
  92. Haeusler AR, Donnelly CJ, Periz G, Simko EAJ, Shaw PG, Kim M-S, Maragakis NJ, Troncoso JC, Pandey A, Sattler R, Rothstein JD, Wang J, 2014. C9orf72 nucleotide repeat structures initiate molecular cascades of disease. Nature; London: 507, 195–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Haidet-Phillips AM, Gross SK, Williams T, Tuteja A, Sherman A, Ko M, Jeong YH, Wong PC, Maragakis NJ, 2013. Altered astrocytic expression of TDP-43 does not influence motor neuron survival. Exp. Neurol 250, 250–259. 10.1016/j.expneurol.2013.10.004 [DOI] [PubMed] [Google Scholar]
  94. Haidet-Phillips AM, Hester ME, Miranda CJ, Meyer K, Braun L, Frakes A, Song S, Likhite S, Murtha MJ, Foust KD, Rao M, Eagle A, Kammesheidt A, Christensen A, Mendell JR, Burghes AHM, Kaspar BK, 2011. Astrocytes from familial and sporadic ALS patients are toxic to motor neurons. Nature Biotechnology 29, 824–828. 10.1038/nbt.1957 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Hall CE, Yao Z, Choi M, Tyzack GE, Serio A, Luisier R, Harley J, Preza E, Arber C, Crisp SJ, Watson PMD, Kullmann DM, Abramov AY, Wray S, Burley R, Loh SHY, Martins LM, Stevens MM, Luscombe NM, Sibley CR, Lakatos A, Ule J, Gandhi S, Patani R, 2017. Progressive Motor Neuron Pathology and the Role of Astrocytes in a Human Stem Cell Model of VCP-Related ALS. Cell Reports 19, 1739–1749. 10.1016/j.celrep.2017.05.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Hansen MK, O’Connor KA, Goehler LE, Watkins LR, Maier SF, 2001. The contribution of the vagus nerve in interleukin-1beta-induced fever is dependent on dose. Am. J. Physiol. Regul. Integr. Comp. Physiol 280, R929–934. 10.1152/ajpregu.2001.280.4.R929 [DOI] [PubMed] [Google Scholar]
  97. Haorah J, Ramirez SH, Schall K, Smith D, Pandya R, Persidsky Y, 2007. Oxidative stress activates protein tyrosine kinase and matrix metalloproteinases leading to blood–brain barrier dysfunction. Journal of Neurochemistry 101, 566–576. 10.1111/j.1471-4159.2006.04393.x [DOI] [PubMed] [Google Scholar]
  98. Harraz MM, Marden JJ, Zhou W, Zhang Y, Williams A, Sharov VS, Nelson K, Luo M, Paulson H, Schöneich C, Engelhardt JF, 2008. SOD1 mutations disrupt redox-sensitive Rac regulation of NADPH oxidase in a familial ALS model. J Clin Invest 118, 659–670. 10.1172/JCI34060 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Hart MP, Gitler AD, 2012. ALS-Associated Ataxin 2 PolyQ Expansions Enhance Stress-Induced Caspase 3 Activation and Increase TDP-43 Pathological Modifications. J. Neurosci 32, 9133–9142. 10.1523/JNEUROSCI.0996-12.2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Hayashi T, Su T-P, 2007. Sigma-1 Receptor Chaperones at the ER-Mitochondrion Interface Regulate Ca2+ Signaling and Cell Survival. Cell 131, 596–610. 10.1016/j.cell.2007.08.036 [DOI] [PubMed] [Google Scholar]
  101. Heemels M-T, 2016. Neurodegenerative diseases. Nature 539, 179 10.1038/539179a [DOI] [PubMed] [Google Scholar]
  102. Heneka MT, Carson MJ, Khoury JE, Landreth GE, Brosseron F, Feinstein DL, Jacobs AH, Wyss-Coray T, Vitorica J, Ransohoff RM, Herrup K, Frautschy SA, Finsen B, Brown GC, Verkhratsky A, Yamanaka K, Koistinaho J, Latz E, Halle A, Petzold GC, Town T, Morgan D, Shinohara ML, Perry VH, Holmes C, Bazan NG, Brooks DJ, Hunot S, Joseph B, Deigendesch N, Garaschuk O, Boddeke E, Dinarello CA, Breitner JC, Cole GM, Golenbock DT, Kummer MP, 2015. Neuroinflammation in Alzheimer’s disease. The Lancet Neurology 14, 388–405. 10.1016/S1474-4422(15)70016-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Higelin J, Catanese A, Semelink-Sedlacek LL, Oeztuerk S, Lutz A-K, Bausinger J, Barbi G, Speit G, Andersen PM, Ludolph AC, Demestre M, Boeckers TM, 2018. NEK1 loss - of-function mutation induces DNA damage accumulation in ALS patient-derived motoneurons. Stem Cell Research 30, 150–162. 10.1016/j.scr.2018.06.005 [DOI] [PubMed] [Google Scholar]
  104. Hindinger C, Bergmann CC, Hinton DR, Phares TW, Parra GI, Hussain S, Savarin C, Atkinson RD, Stohlman SA, 2012. IFN-γ Signaling to Astrocytes Protects from Autoimmune Mediated Neurological Disability. PLOS ONE 7, e42088 10.1371/journal.pone.0042088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Howland DS, Liu J, She Y, Goad B, Maragakis NJ, Kim B, Erickson J, Kulik J, DeVito L, Psaltis G, DeGennaro LJ, Cleveland DW, Rothstein JD, 2002. Focal loss of the glutamate transporter EAAT2 in a transgenic rat model of SOD1 mutant-mediated amyotrophic lateral sclerosis (ALS). Proc Natl Acad Sci U S A 99, 1604–1609. 10.1073/pnas.032539299 [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Hu X, Li P, Guo Y, Wang H, Leak RK, Chen S, Gao Y, Chen J, 2012. Microglia/macrophage polarization dynamics reveal novel mechanism of injury expansion after focal cerebral ischemia. Stroke 43, 3063–3070. 10.1161/STROKEAHA.112.659656 [DOI] [PubMed] [Google Scholar]
  107. Hutten S, Dormann D, 2016. hnRNPA2/B1 Function in Neurodegeneration: It’s a Gain, Not a Loss. Neuron 92, 672–674. 10.1016/j.neuron.2016.11.014 [DOI] [PubMed] [Google Scholar]
  108. Ichiyanagi N, Fujimori K, Yano M, Ishihara-Fujisaki C, Sone T, Akiyama T, Okada Y, Akamatsu W, Matsumoto T, Ishikawa M, Nishimoto Y, Ishihara Y, Sakuma T, Yamamoto T, Tsuiji H, Suzuki N, Warita H, Aoki M, Okano H, 2016. Establishment of In Vitro FUS-Associated Familial Amyotrophic Lateral Sclerosis Model Using Human Induced Pluripotent Stem Cells. Stem Cell Reports 6, 496–510. 10.1016/j.stemcr.2016.02.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Ince P, Stout N, Shaw P, Slade J, Hunziker W, Heizmann CW, Baimbridge KG, 1993. Parvalbumin and calbindin D-28k in the human motor system and in motor neuron disease. Neuropathology and Applied Neurobiology 19, 291–299. 10.1111/j.1365-2990.1993.tb00443.x [DOI] [PubMed] [Google Scholar]
  110. Ince PG, Highley JR, Kirby J, Wharton SB, Takahashi H, Strong MJ, Shaw PJ, 2011. Molecular pathology and genetic advances in amyotrophic lateral sclerosis : an emerging molecular pathway and the significance of glial pathology. Acta Neuropathol 122, 657–671. 10.1007/s00401-011-0913-0 [DOI] [PubMed] [Google Scholar]
  111. Jang J-Y, Cho H, Park H-Y, Rhim H, Kang S, 2017. ALS-linked mutant SOD1 proteins promote Aβ aggregates in ALS through direct interaction with Aβ. Biochemical and Biophysical Research Communications 493, 697–707. 10.1016/j.bbrc.2017.08.127 [DOI] [PubMed] [Google Scholar]
  112. Japtok J, Lojewski X, Naumann M, Klingenstein M, Reinhardt P, Sterneckert J, Putz S, Demestre M, Boeckers TM, Ludolph AC, Liebau S, Storch A, Hermann A, 2015. Stepwise acquirement of hallmark neuropathology in FUS-ALS iPSC models depends on mutation type and neuronal aging. Neurobiology of Disease 82, 420–429. 10.1016/j.nbd.2015.07.017 [DOI] [PubMed] [Google Scholar]
  113. Javed H, Khan MM, Ahmad A, Vaibhav K, Ahmad ME, Khan A, Ashafaq M, Islam F, Siddiqui MS, Safhi MM, Islam F, 2012. Rutin prevents cognitive impairments by ameliorating oxidative stress and neuroinflammation in rat model of sporadic dementia of Alzheimer type. Neuroscience 210, 340–352. 10.1016/j.neuroscience.2012.02.046 [DOI] [PubMed] [Google Scholar]
  114. Jeon GS, Shim Y-M, Lee D-Y, Kim J-S, Kang M, Ahn SH, Shin J-Y, Geum D, Hong YH, Sung J-J, 2018. Pathological Modification of TDP-43 in Amyotrophic Lateral Sclerosis with SOD1 Mutations. Mol Neurobiol 10.1007/s12035-018-1218-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Jin M, Jang E, Suk K, 2014. Lipocalin-2 Acts as a Neuroinflammatogen in Lipopolysaccharide-injected Mice. Exp Neurobiol 23, 155–162. 10.5607/en.2014.23.2.155 [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Johnson BS, Snead D, Lee JJ, McCaffery JM, Shorter J, Gitler AD, 2009. TDP-43 is intrinsically aggregation-prone and ALS-linked mutations accelerate aggregation and increase toxicity. J. Biol. Chem jbc.M109.010264. 10.1074/jbc.M109.010264 [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Johri A, Beal MF, 2012. Mitochondrial Dysfunction in Neurodegenerative Diseases. J Pharmacol Exp Ther 342, 619–630. 10.1124/jpet.112.192138 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Jucker M, Walker LC, 2013. Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature 501, 45–51. 10.1038/nature12481 [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Kaneb HM, Folkmann AW, Belzil VV, Jao L-E, Leblond CS, Girard SL, Daoud H, Noreau A, Rochefort D, Hince P, Szuto A, Levert A, Vidal S, André-Guimont C, Camu W, Bouchard J-P, Dupré N, Rouleau GA, Wente SR, Dion PA, 2015. Deleterious mutations in the essential mRNA metabolism factor, hGle1, in amyotrophic lateral sclerosis. Hum Mol Genet 24, 1363–1373. 10.1093/hmg/ddu545 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Karwoski CJ, Lu H-K, Newman EA, 1989. Spatial Buffering of Light-Evoked Potassium Increases by Retinal Müller (Glial) Cells. Science 244, 578–580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Kawasaki A, Hayashi T, Nakachi K, Trosko JE, Sugihara K, Kotake Y, Ohta S, 2009. Modulation of connexin 43 in rotenone-induced model of Parkinson’s disease. Neuroscience 160, 61–68. 10.1016/j.neuroscience.2009.01.080 [DOI] [PubMed] [Google Scholar]
  122. Kempuraj D, Thangavel R, Natteru P, Selvakumar G, Saeed D, Zahoor H, Zaheer S, Iyer S, Zaheer A, 2016. Neuroinflammation Induces Neurodegeneration. J Neurol Neurosurg Spine 1. [PMC free article] [PubMed] [Google Scholar]
  123. Kenna KP, van Doormaal PTC , Dekker AM, Ticozzi N, Kenna BJ, Diekstra FP, van Rheenen W, Eijk KR, Jones AR, Keagle P, Shatunov A, Sproviero W, Smith BN, van Es MA, Topp SD, Kenna A, Miller JW, Fallini C, Tiloca C, McLaughlin RL, Vance C, Troakes C, Colombrita C, Mora G, Calvo A, Verde F, Al-Sarraj S, King A, Calini D, Belleroche J. de, Baas F, van der Kooi AJ, de Visser M, ten Asbroek ALMA, Sapp PC, McKenna-Yasek D, Polak M, Asress S, Muñoz-Blanco JL, Strom TM, Meitinger T, Morrison KE, Consortium S, D’Alfonso S, Mazzini L, Comi GP, Bo RD, Ceroni M, Gagliardi S, Querin G, Bertolin C, Pensato V, Castellotti B, Corti S, Cereda C, Corrado L, Sorarù G, Lauria G, Williams KL, Leigh PN, Nicholson GA, Blair IP, Leblond CS, Dion PA, Rouleau GA, Pall H, Shaw PJ, Turner MR, Talbot K, Taroni F, Boylan KB, Blitterswijk MV, Rademakers R, Esteban-Pérez J, García-Redondo A, Damme PV, Robberecht W, Chio A, Gellera C, Drepper C, Sendtner M, Ratti A, Glass JD, Mora JS, Basak NA, Hardiman O, Ludolph AC, Andersen PM, Weishaupt JH, Jr, R.H.B., Al-Chalabi A, Silani V, Shaw CE, van den Berg LH, Veldink JH, Landers JE, 2016. NEK1 variants confer susceptibility to amyotrophic lateral sclerosis. Nature Genetics 48, 1037–1042. 10.1038/ng.3626 [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Kigerl KA, Gensel JC, Ankeny DP, Alexander JK, Donnelly DJ, Popovich PG, 2009. Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J. Neurosci 29, 13435–13444. 10.1523/JNEUROSCI.3257-09.2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Kim A, 2014. A Panoramic Overview of Mitochondria and Mitochondrial Redox Biology. Toxicol Res 30, 221–234. 10.5487/TR.2014.30.4.221 [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Kim HJ, Taylor JP, 2017. Lost in Transportation: Nucleocytoplasmic Transport Defects in ALS and Other Neurodegenerative Diseases. Neuron 96, 285–297. 10.1016/j.neuron.2017.07.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Kim J-Y, Jang A, Reddy R, Yoon WH, Jankowsky JL, 2016. Neuronal overexpression of human VAPB slows motor impairment and neuromuscular denervation in a mouse model of ALS. Hum Mol Genet 25, 4661–4673. 10.1093/hmg/ddw294 [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Kim SH, Shanware NP, Bowler MJ, Tibbetts RS, 2010. Amyotrophic Lateral Sclerosis - associated Proteins TDP-43 and FUS/TLS Function in a Common Biochemical Complex to Co-regulate HDAC6 mRNA. J. Biol. Chem 285, 34097–34105. 10.1074/jbc.M110.154831 [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Kimelberg HK, Goderie SK, Higman S, Pang S, Waniewski RA, 1990. Swelling-induced release of glutamate, aspartate, and taurine from astrocyte cultures. J. Neurosci 10, 1583–1591. 10.1523/JNEUROSCI.10-05-01583.1990 [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Kiskinis E, Sandoe J, Williams LA, Boulting GL, Moccia R, Wainger BJ, Han S, Peng T, Thams S, Mikkilineni S, Mellin C, Merkle FT, Davis-Dusenbery BN, Ziller M, Oakley D, Ichida J, Di Costanzo S, Atwater N, Maeder ML, Goodwin MJ, Nemesh J, Handsaker RE, Paull D, Noggle S, McCarroll SA, Joung JK, Woolf CJ, Brown RH, Eggan K, 2014. Pathways Disrupted in Human ALS Motor Neurons Identified through Genetic Correction of Mutant SOD1. Cell Stem Cell 14, 781–795. 10.1016/j.stem.2014.03.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Koppers M, Blokhuis AM, Westeneng H, Terpstra ML, Zundel CAC, Vieira de Sá R, Schellevis RD, Waite AJ, Blake DJ, Veldink JH, van den Berg LH, Pasterkamp RJ, 2015. C9orf72 ablation in mice does not cause motor neuron degeneration or motor deficits. Ann Neurol 78, 426–438. 10.1002/ana.24453 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Krencik R, Weick JP, Liu Y, Zhang Z-J, Zhang S-C, 2011. Specification of transplantable astroglial subtypes from human pluripotent stem cells. Nature Biotechnology 29, 528–534. 10.1038/nbt.1877 [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Kumar A, Stoica BA, Sabirzhanov B, Burns MP, Faden AI, Loane DJ, 2013. Traumatic brain injury in aged animals increases lesion size and chronically alters microglial/macrophage classical and alternative activation states. Neurobiol. Aging 34, 1397–1411. 10.1016/j.neurobiolaging.2012.11.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Kwon I, Xiang S, Kato M, Wu L, Theodoropoulos P, Wang T, Kim J, Yun J, Xie Y, McKnight SL, 2014. Poly-dipeptides encoded by the C9orf72 repeats bind nucleoli, impede RNA biogenesis, and kill cells. Science 345, 1139–1145. 10.1126/science.1254917 [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. La Spada AR, Taylor JP, 2010. Repeat expansion disease: progress and puzzles in disease pathogenesis. Nature Reviews. Genetics; London: 11, 247–58. http://dx.doi.org.ezproxy.cul.columbia.edu/10.1038/nrg2748 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Laflamme N, Lacroix S, Rivest S, 1999. An essential role of interleukin-1beta in mediating NF-kappaB activity and COX-2 transcription in cells of the blood-brain barrier in response to a systemic and localized inflammation but not during endotoxemia. J. Neurosci 19, 10923–10930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Lagier-Tourenne C, Baughn M, Rigo F, Sun S, Liu P, Li H-R, Jiang J, Watt AT, Chun S, Katz M, Qiu J, Sun Y, Ling S-C, Zhu Q, Polymenidou M, Drenner K, Artates JW, McAlonis-Downes M, Markmiller S, Hutt KR, Pizzo DP, Cady J, Harms MB, Baloh RH, Vandenberg SR, Yeo GW, Fu X-D, Bennett CF, Cleveland DW, Ravits J, 2013. Targeted degradation of sense and antisense C9orf72 RNA foci as therapy for ALS and frontotemporal degeneration. Proc. Natl. Acad. Sci. U.S.A 110, E4530–4539. 10.1073/pnas.1318835110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Lagier-Tourenne C, Polymenidou M, Cleveland DW, 2010. TDP-43 and FUS/TLS: emerging roles in RNA processing and neurodegeneration. Hum Mol Genet 19, R46–R64. 10.1093/hmg/ddq137 [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Lee S, Huang EJ, 2017. Modeling ALS and FTD with iPSC-derived neurons. Brain Research, Modeling neurological diseases with patient-derived neurons 1656, 88–97. 10.1016/j.brainres.2015.10.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Lee S, Park J-Y, Lee W-H, Kim H, Park H-C, Mori K, Suk K, 2009. Lipocalin-2 Is an Autocrine Mediator of Reactive Astrocytosis. J. Neurosci 29, 234–249. 10.1523/JNEUROSCI.5273-08.2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Lenzi J, Pagani F, De Santis R, Limatola C, Bozzoni I, Di Angelantonio S, Rosa A, 2016. Differentiation of control and ALS mutant human iPSCs into functional skeletal muscle cells, a tool for the study of neuromuscolar diseases. Stem Cell Research 17, 140–147. 10.1016/j.scr.2016.06.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Lenzi J, Santis RD, Turris V. de, Morlando M, Laneve P, Calvo A, Caliendo V, Chiò A, Rosa A, Bozzoni I, 2015. ALS mutant FUS proteins are recruited into stress granules in induced pluripotent stem cell-derived motoneurons. Disease Models & Mechanisms 8, 755–766. 10.1242/dmm.020099 [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Levine JB, Kong J, Nadler M, Xu Z, 1999. Astrocytes interact intimately with degenerating motor neurons in mouse amyotrophic lateral sclerosis (ALS). Glia 28, 215–224. [DOI] [PubMed] [Google Scholar]
  144. Li TM, Alberman E, Swash M, 1988. Comparison of sporadic and familial disease amongst 580 cases of motor neuron disease. Journal of Neurology, Neurosurgery & Psychiatry 51, 778–784. 10.1136/jnnp.51.6.778 [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Li Y, Balasubramanian U, Cohen D, Zhang P-W, Mosmiller E, Sattler R, Maragakis NJ, Rothstein JD, 2015. A Comprehensive Library of Familial Human Amyotrophic Lateral Sclerosis Induced Pluripotent Stem Cells. PLOS ONE 10, e0118266 10.1371/journal.pone.0118266 [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, Bennett ML, Münch AE, Chung W-S, Peterson TC, Wilton DK, Frouin A, Napier BA, Panicker N, Kumar M, Buckwalter MS, Rowitch DH, Dawson VL, Dawson TM, Stevens B, Barres BA, 2017. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541, 481–487. 10.1038/nature21029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Linker RA, Lee D-H, Demir S, Wiese S, Kruse N, Siglienti I, Gerhardt E, Neumann H, Sendtner M, Lühder F, Gold R, 2010. Functional role of brain-derived neurotrophic factor in neuroprotective autoimmunity: therapeutic implications in a model of multiple sclerosis. Brain 133, 2248–2263. 10.1093/brain/awq179 [DOI] [PubMed] [Google Scholar]
  148. Lino MM, Schneider C, Caroni P, 2002. Accumulation of SOD1 Mutants in Postnatal Motoneurons Does Not Cause Motoneuron Pathology or Motoneuron Disease. J. Neurosci 22, 4825–4832. 10.1523/JNEUROSCI.22-12-04825.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Liu J, Hu J, Ludlow AT, Pham JT, Shay JW, Rothstein JD, Corey DR, 2017. c9orf72 Disease-Related Foci Are Each Composed of One Mutant Expanded Repeat RNA. Cell Chemical Biology 24, 141–148. 10.1016/j.chembiol.2016.12.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Liu J, Lillo C, Jonsson PA, Velde CV, Ward CM, Miller TM, Subramaniam JR, Rothstein JD, Marklund S, Andersen PM, Brännström T, Gredal O, Wong PC, Williams DS, Cleveland DW, 2004. Toxicity of Familial ALS-Linked SOD1 Mutants from Selective Recruitment to Spinal Mitochondria. Neuron 43, 5–17. 10.1016/j.neuron.2004.06.016 [DOI] [PubMed] [Google Scholar]
  151. Liu M-L, Zang T, Zhang C-L, 2016. Direct Lineage Reprogramming Reveals Disease-Specific Phenotypes of Motor Neurons from Human ALS Patients. Cell Reports 14, 115–128. 10.1016/j.celrep.2015.12.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Lopez-Gonzalez R, Lu Y, Gendron TF, Karydas A, Tran H, Yang D, Petrucelli L, Miller BL, Almeida S, Gao F-B, 2016. Poly(GR) in C9ORF72-Related ALS/FTD Compromises Mitochondrial Function and Increases Oxidative Stress and DNA Damage in iPSC-Derived Motor Neurons. Neuron 92, 383–391. 10.1016/j.neuron.2016.09.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Ludtmann MHR, Arber C, Bartolome F, Vicente M. de, Preza E, Carro E, Houlden H, Gandhi S, Wray S, Abramov AY, 2017. Mutations in valosin-containing protein (VCP) decrease ADP/ATP translocation across the mitochondrial membrane and impair energy metabolism in human neurons. J. Biol. Chem 292, 8907–8917. 10.1074/jbc.M116.762898 [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Mackenzie IR, Nicholson AM, Sarkar M, Messing J, Purice MD, Pottier C, Annu K, Baker M, Perkerson RB, Kurti A, Matchett BJ, Mittag T, Temirov J, Hsiung G-YR, Krieger C, Murray ME, Kato M, Fryer JD, Petrucelli L, Zinman L, Weintraub S, Mesulam M, Keith J, Zivkovic SA, Hirsch-Reinshagen V, Roos RP, Züchner S, Graff-Radford NR, Petersen RC, Caselli RJ, Wszolek ZK, Finger E, Lippa C, Lacomis D, Stewart H, Dickson DW, Kim HJ, Rogaeva E, Bigio E, Boylan KB, Taylor JP, Rademakers R, 2017. TIA1 Mutations in Amyotrophic Lateral Sclerosis and Frontotemporal Dementia Promote Phase Separation and Alter Stress Granule Dynamics. Neuron 95, 808–816.e9. 10.1016/j.neuron.2017.07.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Mackenzie IRA, Bigio EH, Ince PG, Geser F, Neumann M, Cairns NJ, Kwong LK, Forman MS, Ravits J, Stewart H, Eisen A, McClusky L, Kretzschmar HA, Monoranu CM, Highley JR, Kirby J, Siddique T, Shaw PJ, Lee VM-Y, Trojanowski JQ, 2007. Pathological TDP-43 distinguishes sporadic amyotrophic lateral sclerosis from amyotrophic lateral sclerosis with SOD1 mutations. Annals of Neurology 61, 427–434. 10.1002/ana.21147 [DOI] [PubMed] [Google Scholar]
  156. Madill M, McDonagh K, Ma J, Vajda A, McLoughlin P, O’Brien T, Hardiman O, Shen S, 2017. Amyotrophic lateral sclerosis patient iPSC-derived astrocytes impair autophagy via non-cell autonomous mechanisms. Molecular Brain 10, 22 10.1186/s13041-017-0300-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Maniatis S, Äijö T, Vickovic S, Braine C, Kang K, Mollbrink A, Fagegaltier D, Andrusivová Ž, Saarenpää S, Saiz-Castro G, Cuevas M, Watters A, Lundeberg J, Bonneau R, Phatnani H, 2019. Spatiotemporal dynamics of molecular pathology in amyotrophic lateral sclerosis. Science 364, 89–93. 10.1126/science.aav9776 [DOI] [PubMed] [Google Scholar]
  158. Marchetto MCN, Muotri AR, Mu Y, Smith AM, Cezar GG, Gage FH, 2008. Non-Cell-Autonomous Effect of Human SOD1G37R Astrocytes on Motor Neurons Derived from Human Embryonic Stem Cells. Cell Stem Cell 3, 649–657. 10.1016/j.stem.2008.10.001 [DOI] [PubMed] [Google Scholar]
  159. Marden JJ, Harraz MM, Williams AJ, Nelson K, Luo M, Paulson H, Engelhardt JF, 2007. Redox modifier genes in amyotrophic lateral sclerosis in mice. J. Clin. Invest 117, 2913–2919. 10.1172/JCI31265 [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Maurel C, Dangoumau A, Marouillat S, Brulard C, Chami A, Hergesheimer R, Corcia P, Blasco H, Andres CR, Vourc’h P, 2018. Causative Genes in Amyotrophic Lateral Sclerosis and Protein Degradation Pathways: a Link to Neurodegeneration. Mol Neurobiol 1–20. 10.1007/s12035-017-0856-0 [DOI] [PubMed] [Google Scholar]
  161. McCann SK, Roulston CL, 2013. NADPH Oxidase as a Therapeutic Target for Neuroprotection against Ischaemic Stroke: Future Perspectives. Brain Sci 3, 561–598. 10.3390/brainsci3020561 [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. McCoy MK, Martinez TN, Ruhn KA, Szymkowski DE, Smith CG, Botterman BR, Tansey KE, Tansey MG, 2006. Blocking soluble tumor necrosis factor signaling with dominant-negative tumor necrosis factor inhibitor attenuates loss of dopaminergic neurons in models of Parkinson’s disease. J. Neurosci 26, 9365–9375. 10.1523/JNEUROSCI.1504-06.2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Meeuwsen S, Persoon‐Deen C, Bsibsi M, Ravid R, Noort JMV, 2003. Cytokine, chemokine and growth factor gene profiling of cultured human astrocytes after exposure to proinflammatory stimuli. Glia 43, 243–253. 10.1002/glia.10259 [DOI] [PubMed] [Google Scholar]
  164. Meyer K, Ferraiuolo L, Miranda CJ, Likhite S, McElroy S, Renusch S, Ditsworth D, Lagier-Tourenne C, Smith RA, Ravits J, Burghes AH, Shaw PJ, Cleveland DW, Kolb SJ, Kaspar BK, 2014. Direct conversion of patient fibroblasts demonstrates non-cell autonomous toxicity of astrocytes to motor neurons in familial and sporadic ALS. PNAS 111, 829–832. 10.1073/pnas.1314085111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Miller JD, Ganat YM, Kishinevsky S, Bowman RL, Liu B, Tu EY, Mandal P, Vera E, Shim J, Kriks S, Taldone T, Fusaki N, Tomishima MJ, Krainc D, Milner TA, Rossi DJ, Studer L, 2013. Human iPSC-based Modeling of Late-Onset Disease via Progerin-induced Aging. Cell Stem Cell 13, 691–705. 10.1016/j.stem.2013.11.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Mitchell J, Paul P, Chen H-J, Morris A, Payling M, Falchi M, Habgood J, Panoutsou S, Winkler S, Tisato V, Hajitou A, Smith B, Vance C, Shaw C, Mazarakis ND, Belleroche J. de, 2010. Familial amyotrophic lateral sclerosis is associated with a mutation in D-amino acid oxidase. PNAS 107, 7556–7561. 10.1073/pnas.0914128107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Mitne-Neto M, Machado-Costa M, Marchetto MCN, Bengtson MH, Joazeiro CA, Tsuda H, Bellen HJ, Silva HCA, Oliveira ASB, Lazar M, Muotri AR, Zatz M, 2011. Downregulation of VAPB expression in motor neurons derived from induced pluripotent stem cells of ALS8 patients. Hum Mol Genet 20, 3642–3652. 10.1093/hmg/ddr284 [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Morello G, Spampinato AG, Cavallaro S, 2017. Neuroinflammation and ALS: Transcriptomic Insights into Molecular Disease Mechanisms and Therapeutic Targets. Mediators of Inflammation 10.1155/2017/7070469 [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Morgan S, Shoai M, Fratta P, Sidle K, Orrell R, Sweeney MG, Shatunov A, Sproviero W, Jones A, Al-Chalabi A, Malaspina A, Houlden H, Hardy J, Pittman A, 2015. Investigation of next-generation sequencing technologies as a diagnostic tool for amyotrophic lateral sclerosis. Neurobiology of Aging 36, 1600.e5–1600.e8. 10.1016/j.neurobiolaging.2014.12.017 [DOI] [PubMed] [Google Scholar]
  170. Mori K, Weng S-M, Arzberger T, May S, Rentzsch K, Kremmer E, Schmid B, Kretzschmar HA, Cruts M, Broeckhoven CV, Haass C, Edbauer D, 2013. The C9orf72 GGGGCC Repeat Is Translated into Aggregating Dipeptide-Repeat Proteins in FTLD/ALS. Science 339, 1335–1338. 10.1126/science.1232927 [DOI] [PubMed] [Google Scholar]
  171. Nagai M, Re DB, Nagata T, Chalazonitis A, Jessell TM, Wichterle H, Przedborski S, 2007. Astrocytes expressing ALS-linked mutated SOD1 release factors selectively toxic to motor neurons. Nat Neurosci 10, 615–622. 10.1038/nn1876 [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Nakamura R, Sone J, Atsuta N, Tohnai G, Watanabe Hazuki, Yokoi D, Nakatochi M, Watanabe Hirohisa, Ito M, Senda J, Katsuno M, Tanaka F, Li Y, Izumi Y, Morita M, Taniguchi A, Kano O, Oda M, Kuwabara S, Abe K, Aiba I, Okamoto K, Mizoguchi K, Hasegawa K, Aoki M, Hattori N, Tsuji S, Nakashima K, Kaji R, Sobue G, 2016. Next-generation sequencing of 28 ALS-related genes in a Japanese ALS cohort. Neurobiology of Aging 39, 219.e1–219.e8. 10.1016/j.neurobiolaging.2015.11.030 [DOI] [PubMed] [Google Scholar]
  173. Naumann M, Pal A, Goswami A, Lojewski X, Japtok J, Vehlow A, Naujock M, Günther R, Jin M, Stanslowsky N, Reinhardt P, Sterneckert J, Frickenhaus M, Pan-Montojo F, Storkebaum E, Poser I, Freischmidt A, Weishaupt JH, Holzmann K, Troost D, Ludolph AC, Boeckers TM, Liebau S, Petri S, Cordes N, Hyman AA, Wegner F, Grill SW, Weis J, Storch A, Hermann A, 2018. Impaired DNA damage response signaling by FUS-NLS mutations leads to neurodegeneration and FUS aggregate formation. Nat Commun 9, 335 10.1038/s41467-017-02299-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Nihei Y, Ito D, Suzuki N, 2012. Roles of Ataxin-2 in Pathological Cascades Mediated by TAR DNA-binding Protein 43 (TDP-43) and Fused in Sarcoma (FUS). J. Biol. Chem 287, 41310–41323. 10.1074/jbc.M112.398099 [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Nishihira Y, Tan C-F, Onodera O, Toyoshima Y, Yamada M, Morita T, Nishizawa M, Kakita A, Takahashi H, 2008. Sporadic amyotrophic lateral sclerosis: two pathological patterns shown by analysis of distribution of TDP-43-immunoreactive neuronal and glial cytoplasmic inclusions. Acta Neuropathol 116, 169 10.1007/s00401-008-0385-z [DOI] [PubMed] [Google Scholar]
  176. Nobuta H, Ghiani CA, Paez PM, Spreuer V, Dong H, Korsak RA, Manukyan A, Li J, Vinters HV, Huang EJ, Rowitch DH, Sofroniew MV, Campagnoni AT, Vellis J. de, Waschek JA, 2012. STAT3-Mediated astrogliosis protects myelin development in neonatal brain injury. Annals of Neurology 72, 750–765. 10.1002/ana.23670 [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Nonaka T, Masuda-Suzukake M, Arai T, Hasegawa Y, Akatsu H, Obi T, Yoshida M, Murayama S, Mann DMA, Akiyama H, Hasegawa M, 2013. Prion-like Properties of Pathological TDP-43 Aggregates from Diseased Brains. Cell Reports 4, 124–134. 10.1016/j.celrep.2013.06.007 [DOI] [PubMed] [Google Scholar]
  178. Norden DM, Trojanowski PJ, Villanueva E, Navarro E, Godbout JP, 2016. Sequential Activation of Microglia and Astrocyte Cytokine Expression Precedes Increased Iba-1 or GFAP Immunoreactivity following Systemic Immune Challenge. Glia 64, 300–316. 10.1002/glia.22930 [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Oberheim NA, Goldman SA, Nedergaard M, 2012. Heterogeneity of Astrocytic Form and Function, in: Milner R (Ed.), Astrocytes: Methods and Protocols, Methods in Molecular Biology Humana Press, Totowa, NJ, pp. 23–45. 10.1007/978-1-61779-452-0_3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Oberheim NA, Takano T, Han X, He W, Lin JHC, Wang F, Xu Q, Wyatt JD, Pilcher W, Ojemann JG, Ransom BR, Goldman SA, Nedergaard M, 2009. Uniquely Hominid Features of Adult Human Astrocytes. J. Neurosci 29, 3276–3287. 10.1523/JNEUROSCI.4707-08.2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Oeckl P, Weydt P, Steinacker P, Anderl-Straub S, Nordin F, Volk AE, Diehl-Schmid J, Andersen PM, Kornhuber J, Danek A, Fassbender K, Fliessbach K, Degeneration, G.C. for F.L., Jahn H, Lauer M, Müller K, Knehr A, Prudlo J, Schneider A, Thal DR, Yilmazer-Hanke D, Weishaupt JH, Ludolph AC, Otto M, 2018. Different neuroinflammatory profile in amyotrophic lateral sclerosis and frontotemporal dementia is linked to the clinical phase. J Neurol Neurosurg Psychiatry jnnp-2018–318868. 10.1136/jnnp-2018-318868 [DOI] [PubMed] [Google Scholar]
  182. Orellana Juan A., Froger Nicolas, Ezan Pascal, Jiang Jean X., Bennett Michael V. L., Naus Christian C., Giaume Christian, Sáez Juan C., 2011. ATP and glutamate released via astroglial connexin 43 hemichannels mediate neuronal death through activation of pannexin 1 hemichannels. Journal of Neurochemistry 118, 826–840. 10.1111/j.1471-4159.2011.07210.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. O’Rourke JG, Bogdanik L, Yáñez A, Lall D, Wolf AJ, Muhammad AKMG, Ho R, Carmona S, Vit JP, Zarrow J, Kim KJ, Bell S, Harms MB, Miller TM, Dangler CA, Underhill DM, Goodridge HS, Lutz CM, Baloh RH, 2016. C9orf72 is required for proper macrophage and microglial function in mice. Science 351, 1324–1329. 10.1126/science.aaf1064 [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Pal A, Fontanilla D, Gopalakrishnan A, Chae Y-K, Markley JL, Ruoho AE, 2012. The sigma-1 receptor protects against cellular oxidative stress and activates antioxidant response elements. European Journal of Pharmacology 682, 12–20. 10.1016/j.ejphar.2012.01.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Park KW, Baik HH, Jin BK, 2009. IL-13-Induced Oxidative Stress via Microglial NADPH Oxidase Contributes to Death of Hippocampal Neurons In Vivo. The Journal of Immunology 183, 4666–4674. 10.4049/jimmunol.0803392 [DOI] [PubMed] [Google Scholar]
  186. Pehar Mariana, Cassina Patricia, Vargas Marcelo R., Castellanos Raquel, Viera Liliana, Beckman Joseph S., Estévez Alvaro G., Barbeito Luis, 2004. Astrocytic production of nerve growth factor in motor neuron apoptosis: implications for amyotrophic lateral sclerosis. Journal of Neurochemistry 89, 464–473. 10.1111/j.1471-4159.2004.02357.x [DOI] [PubMed] [Google Scholar]
  187. Pelegrini AL, Moura DJ, Brenner BL, Ledur PF, Maques GP, Henriques JAP, Saffi J, Lenz G, 2010. Nek1 silencing slows down DNA repair and blocks DNA damage-induced cell cycle arrest. Mutagenesis 25, 447–454. 10.1093/mutage/geq026 [DOI] [PubMed] [Google Scholar]
  188. Perrone F, Nguyen HP, Van Mossevelde S, Moisse M, Sieben A, Santens P, De Bleecker J, Vandenbulcke M, Engelborghs S, Baets J, Cras P, Vandenberghe R, De Jonghe P, De Deyn PP, Martin J-J, Van Damme P, Van Broeckhoven C, van der Zee J, Belgian Neurology consortium, 2017. Investigating the role of ALS genes CHCHD10 and TUBA4A in Belgian FTD-ALS spectrum patients. Neurobiol. Aging 51, 177.e9–177.e16. 10.1016/j.neurobiolaging.2016.12.008 [DOI] [PubMed] [Google Scholar]
  189. Phatnani HP, Guarnieri P, Friedman BA, Carrasco MA, Muratet M, O’Keeffe S, Nwakeze C, Pauli-Behn F, Newberry KM, Meadows SK, Tapia JC, Myers RM, Maniatis T, 2013. Intricate interplay between astrocytes and motor neurons in ALS. Proc Natl Acad Sci U S A 110, E756–E765. 10.1073/pnas.1222361110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Pintado C, Macías S, Domínguez-Martín H, Castaño A, Ruano D, 2017. Neuroinflammation alters cellular proteostasis by producing endoplasmic reticulum stress, autophagy activation and disrupting ERAD activation. Scientific Reports 7, 8100 10.1038/s41598-017-08722-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Porter JT, McCarthy KD, 1996. Hippocampal Astrocytes In Situ Respond to Glutamate Released from Synaptic Terminals. J. Neurosci 16, 5073–5081. 10.1523/JNEUROSCI.16-16-05073.1996 [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Pramatarova A, Laganière J, Roussel J, Brisebois K, Rouleau GA, 2001. Neuron-specific expression of mutant superoxide dismutase 1 in transgenic mice does not lead to motor impairment. J. Neurosci 21, 3369–3374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Purandare N, Somayajulu M, Hüttemann M, Grossman LI, Aras S, 2018. The cellular stress proteins CHCHD10 and MNRR1 (CHCHD2): Partners in mitochondrial and nuclear function and dysfunction. J. Biol. Chem 293, 6517–6529. 10.1074/jbc.RA117.001073 [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Purice MD, Taylor JP, 2018. Linking hnRNP Function to ALS and FTD Pathology. Front Neurosci 12, 326 10.3389/fnins.2018.00326 [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Qian K, Huang H, Peterson A, Hu B, Maragakis NJ, Ming G, Chen H, Zhang S-C, 2017. Sporadic ALS Astrocytes Induce Neuronal Degeneration In Vivo. Stem Cell Reports 8, 843–855. 10.1016/j.stemcr.2017.03.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Qosa H, Lichter J, Sarlo M, Markandaiah SS, McAvoy K, Richard J-P, Jablonski MR, Maragakis NJ, Pasinelli P, Trotti D, 2016. Astrocytes drive upregulation of the multidrug resistance transporter ABCB1 (P-Glycoprotein) in endothelial cells of the blood-brain barrier in mutant superoxide dismutase 1-linked amyotrophic lateral sclerosis. Glia 64, 1298–1313. 10.1002/glia.23003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Ravaioli F, Bacalini MG, Franceschi C, Garagnani P, 2018. Age-Related Epigenetic Derangement upon Reprogramming and Differentiation of Cells from the Elderly. Genes 9, 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Renton AE, Majounie E, Waite A, Simón-Sánchez J, Rollinson S, Gibbs JR, Schymick JC, Laaksovirta H, van Swieten JC, Myllykangas L, Kalimo H, Paetau A, Abramzon Y, Remes AM, Kaganovich A, Scholz SW, Duckworth J, Ding J, Harmer DW, Hernandez DG, Johnson JO, Mok K, Ryten M, Trabzuni D, Guerreiro RJ, Orrell RW, Neal J, Murray A, Pearson J, Jansen IE, Sondervan D, Seelaar H, Blake D, Young K, Halliwell N, Callister JB, Toulson G, Richardson A, Gerhard A, Snowden J, Mann D, Neary D, Nalls MA, Peuralinna T, Jansson L, Isoviita V-M, Kaivorinne A-L, Hölttä-Vuori M, Ikonen E, Sulkava R, Benatar M, Wuu J, Chiò A, Restagno G, Borghero G, Sabatelli M, ITALSGEN Consortium, Heckerman D, Rogaeva E, Zinman L, Rothstein JD, Sendtner M, Drepper C, Eichler EE, Alkan C, Abdullaev Z, Pack SD, Dutra A, Pak E, Hardy J, Singleton A, Williams NM, Heutink P, Pickering-Brown S, Morris HR, Tienari PJ, Traynor BJ, 2011. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 72, 257–268. 10.1016/j.neuron.2011.09.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  199. Robberecht W, Philips T, 2013. The changing scene of amyotrophic lateral sclerosis. Nature Reviews Neuroscience 14, 248–264. 10.1038/nrn3430 [DOI] [PubMed] [Google Scholar]
  200. Rothstein JD, Martin LJ, Kuncl RW, 1992. Decreased glutamate transport by the brain and spinal cord in amyotrophic lateral sclerosis. N. Engl. J. Med 326, 1464–1468. 10.1056/NEJM199205283262204 [DOI] [PubMed] [Google Scholar]
  201. Rothstein JD, Van Kammen M, Levey AI, Martin LJ, Kuncl RW, 1995. Selective loss of glial glutamate transporter GLT-1 in amyotrophic lateral sclerosis. Ann. Neurol 38, 73–84. 10.1002/ana.410380114 [DOI] [PubMed] [Google Scholar]
  202. Rudnick ND, Griffey CJ, Guarnieri P, Gerbino V, Wang X, Piersaint JA, Tapia JC, Rich MM, Maniatis T, 2017. Distinct roles for motor neuron autophagy early and late in the SOD1G93A mouse model of ALS. Proc. Natl. Acad. Sci. U.S.A 114, E8294–E8303. 10.1073/pnas.1704294114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. Ryter SW, Kim HP, Hoetzel A, Park JW, Nakahira K, Wang X, Choi AMK, 2007. Mechanisms of cell death in oxidative stress. Antioxid. Redox Signal 9, 49–89. 10.1089/ars.2007.9.49 [DOI] [PubMed] [Google Scholar]
  204. Said Ahmed M, Hung WY, Zu JS, Hockberger P, Siddique T, 2000. Increased reactive oxygen species in familial amyotrophic lateral sclerosis with mutations in SOD1. J. Neurol. Sci 176, 88–94. [DOI] [PubMed] [Google Scholar]
  205. Santello M, Bezzi P, Volterra A, 2011. TNFα Controls Glutamatergic Gliotransmission in the Hippocampal Dentate Gyrus. Neuron 69, 988–1001. 10.1016/j.neuron.2011.02.003 [DOI] [PubMed] [Google Scholar]
  206. Santos R, Vadodaria KC, Jaeger BN, Mei A, Lefcochilos-Fogelquist S, Mendes APD, Erikson G, Shokhirev M, Randolph-Moore L, Fredlender C, Dave S, Oefner R, Fitzpatrick C, Pena M, Barron JJ, Ku M, Denli AM, Kerman BE, Gage FH, 2017. Differentiation of Inflammation-Responsive Astrocytes from Glial Progenitors Generated from Human Induced Pluripotent Stem Cells. Stem Cell Reports 8, 1757–1769. 10.1016/j.stemcr.2017.05.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Sareen D, O’Rourke JG, Meera P, Muhammad AKMG, Grant S, Simpkinson M, Bell S, Carmona S, Ornelas L, Sahabian A, Gendron T, Petrucelli L, Baughn M, Ravits J, Harms MB, Rigo F, Bennett CF, Otis TS, Svendsen CN, Baloh RH, 2013. Targeting RNA Foci in iPSC-Derived Motor Neurons from ALS Patients with a C9ORF72 Repeat Expansion. Science Translational Medicine 5, 208ra149–208ra149. 10.1126/scitranslmed.3007529 [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Sasabe J, Chiba T, Yamada M, Okamoto K, Nishimoto I, Matsuoka M, Aiso S, 2007. D-Serine is a key determinant of glutamate toxicity in amyotrophic lateral sclerosis. EMBO J 26, 4149–4159. 10.1038/sj.emboj.7601840 [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Sasabe J, Miyoshi Y, Suzuki M, Mita M, Konno R, Matsuoka M, Hamase K, Aiso S, 2012. d-Amino acid oxidase controls motoneuron degeneration through d-serine. PNAS 109, 627–632. 10.1073/pnas.1114639109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Sato K, Otomo A, Ueda MT, Hiratsuka Y, Suzuki-Utsunomiya K, Sugiyama J, Murakoshi S, Mitsui S, Ono S, Nakagawa S, Shang H-F, Hadano S, 2018. Altered oligomeric states in pathogenic ALS2 variants associated with juvenile motor neuron diseases cause loss of ALS2-mediated endosomal function. J. Biol. Chem jbc.RA118.003849. 10.1074/jbc.RA118.003849 [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Schipke CG, Kettenmann H, 2004. Astrocyte responses to neuronal activity. Glia 47, 226–232. 10.1002/glia.20029 [DOI] [PubMed] [Google Scholar]
  212. Schitine C, Nogaroli L, Costa MR, Hedin-Pereira C, 2015. Astrocyte heterogeneity in the brain: from development to disease. Front Cell Neurosci 9 10.3389/fncel.2015.00076 [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Schmitt ND, Agar JN, 2017. Parsing disease-relevant protein modifications from epiphenomena: perspective on the structural basis of SOD1-mediated ALS. Journal of Mass Spectrometry 52, 480–491. 10.1002/jms.3953 [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Schreiner B, Romanelli E, Liberski P, Ingold-Heppner B, Sobottka-Brillout B, Hartwig T, Chandrasekar V, Johannssen H, Zeilhofer HU, Aguzzi A, Heppner F, Kerschensteiner M, Becher B, 2015. Astrocyte Depletion Impairs Redox Homeostasis and Triggers Neuronal Loss in the Adult CNS. Cell Reports 12, 1377–1384. 10.1016/j.celrep.2015.07.051 [DOI] [PubMed] [Google Scholar]
  215. Schwartz JC, Podell ER, Han SSW, Berry JD, Eggan KC, Cech TR, Strome S, 2014. FUS is sequestered in nuclear aggregates in ALS patient fibroblasts. MBoC 25, 2571–2578. 10.1091/mbc.e14-05-1007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Scotter EL, Chen H-J, Shaw CE, 2015. TDP-43 Proteinopathy and ALS: Insights into Disease Mechanisms and Therapeutic Targets. Neurotherapeutics 12, 352–363. 10.1007/s13311-015-0338-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Sellier C, Campanari M-L, Corbier CJ, Gaucherot A, Kolb‐Cheynel I, Oulad‐Abdelghani M, Ruffenach F, Page A, Ciura S, Kabashi E, Charlet‐Berguerand N, 2016. Loss of C9ORF72 impairs autophagy and synergizes with polyQ Ataxin-2 to induce motor neuron dysfunction and cell death. The EMBO Journal 35, 1276–1297. 10.15252/embj.201593350 [DOI] [PMC free article] [PubMed] [Google Scholar]
  218. Selvaraj BT, Livesey MR, Chandran S, 2017. Modeling the C9ORF72 repeat expansion mutation using human induced pluripotent stem cells. Brain Pathology 27, 518–524. 10.1111/bpa.12520 [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Selvaraj BT, Livesey MR, Zhao C, Gregory JM, James OT, Cleary EM, Chouhan AK, Gane AB, Perkins EM, Dando O, Lillico SG, Lee Y-B, Nishimura AL, Poreci U, Thankamony S, Pray M, Vasistha NA, Magnani D, Borooah S, Burr K, Story D, McCampbell A, Shaw CE, Kind PC, Aitman TJ, Whitelaw CBA, Wilmut I, Smith C, Miles GB, Hardingham GE, Wyllie DJA, Chandran S, 2018. C9ORF72 repeat expansion causes vulnerability of motor neurons to Ca 2+ -permeable AMPA receptor-mediated excitotoxicity. Nature Communications 9, 347 10.1038/s41467-017-02729-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  220. Seminary ER, Sison SL, Ebert AD, 2018. Modeling Protein Aggregation and the Heat Shock Response in ALS iPSC-Derived Motor Neurons. Front. Neurosci 12 10.3389/fnins.2018.00086 [DOI] [PMC free article] [PubMed] [Google Scholar]
  221. Serio A, Bilican B, Barmada SJ, Ando DM, Zhao C, Siller R, Burr K, Haghi G, Story D, Nishimura AL, Carrasco MA, Phatnani HP, Shum C, Wilmut I, Maniatis T, Shaw CE, Finkbeiner S, Chandran S, 2013. Astrocyte pathology and the absence of non-cell autonomy in an induced pluripotent stem cell model of TDP-43 proteinopathy. Proc. Natl. Acad. Sci. U.S.A 110, 4697–4702. 10.1073/pnas.1300398110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  222. Serio A, Patani R, 2018. Concise Review: The Cellular Conspiracy of Amyotrophic Lateral Sclerosis. Stem Cells 36, 293–303. 10.1002/stem.2758 [DOI] [PubMed] [Google Scholar]
  223. Serrats J, Schiltz JC, García-Bueno B, van Rooijen N, Reyes TM, Sawchenko PE, 2010. Dual roles for perivascular macrophages in immune-to-brain signaling. Neuron 65, 94–106. 10.1016/j.neuron.2009.11.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Shaltouki Atossa, Peng Jun, Liu Qiuyue, Rao Mahendra S., Zeng Xianmin, 2013. Efficient Generation of Astrocytes from Human Pluripotent Stem Cells in Defined Conditions. STEM CELLS 31, 941–952. 10.1002/stem.1334 [DOI] [PubMed] [Google Scholar]
  225. Shaw Pamela J., Forrest V, Ince PG, Richardson JP, Wastell HJ, 1995. CSF and Plasma Amino Acid Levels in Motor Neuron Disease: Elevation of CSF Glutamate in a Subset of Patients. Neurodegeneration 4, 209–216. 10.1006/neur.1995.0026 [DOI] [PubMed] [Google Scholar]
  226. Shaw PJ, Ince PG, Falkous G, Mantle D, 1995. Oxidative damage to protein in sporadic motor neuron disease spinal cord. Ann. Neurol 38, 691–695. 10.1002/ana.410380424 [DOI] [PubMed] [Google Scholar]
  227. Shibata N, Nagai R, Uchida K, Horiuchi S, Yamada S, Hirano A, Kawaguchi M, Yamamoto T, Sasaki S, Kobayashi M, 2001. Morphological evidence for lipid peroxidation and protein glycoxidation in spinal cords from sporadic amyotrophic lateral sclerosis patients. Brain Res 917, 97–104. [DOI] [PubMed] [Google Scholar]
  228. Shinoda Y, Tagashira H, Fukunaga K, 2015. ALS-related SIGMAR1 Missense Mutation Causes TDP-43 Inclusion and Mitochondrial Injury. The FASEB Journal 29, 564.3. 10.1096/fasebj.29.1_supplement.564.3 [DOI] [Google Scholar]
  229. Shodai A, Morimura T, Ido A, Uchida T, Ayaki T, Takahashi Rina, Kitazawa S, Suzuki S, Shirouzu M, Kigawa T, Muto Y, Yokoyama S, Takahashi Ryosuke, Kitahara R, Ito H, Fujiwara N, Urushitani M, 2013. Aberrant Assembly of RNA Recognition Motif 1 Links to Pathogenic Conversion of TAR DNA-binding Protein of 43 kDa (TDP-43). J. Biol. Chem 288, 14886–14905. 10.1074/jbc.M113.451849 [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. Simpson EP, Henry YK, Henkel JS, Smith RG, Appel SH, 2004. Increased lipid peroxidation in sera of ALS patients: a potential biomarker of disease burden. Neurology 62, 1758–1765. [DOI] [PubMed] [Google Scholar]
  231. Son EY, Ichida JK, Wainger BJ, Toma JS, Rafuse VF, Woolf CJ, Eggan K, 2011. Conversion of Mouse and Human Fibroblasts into Functional Spinal Motor Neurons. Cell Stem Cell 9, 205–218. 10.1016/j.stem.2011.07.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Song E, Fan P, Huang B, Deng H, Cheung BMY, Félétou M, Vilaine J, Villeneuve N, Xu A, Vanhoutte PM, Wang Y, 2014. Deamidated Lipocalin‐2 Induces Endothelial Dysfunction and Hypertension in Dietary Obese Mice. J Am Heart Assoc 3 10.1161/JAHA.114.000837 [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Spang N, Feldmann A, Huesmann H, Bekbulat F, Schmitt V, Hiebel C, Koziollek-Drechsler I, Clement AM, Moosmann B, Jung J, Behrends C, Dikic I, Kern A, Behl C, 2014. RAB3GAP1 and RAB3GAP2 modulate basal and rapamycin-induced autophagy. Autophagy 10, 2297–2309. 10.4161/15548627.2014.994359 [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Spreux-Varoquaux O, Bensimon G, Lacomblez L, Salachas F, Pradat PF, Le Forestier N, Marouan A, Dib M, Meininger V, 2002. Glutamate levels in cerebrospinal fluid in amyotrophic lateral sclerosis: a reappraisal using a new HPLC method with coulometric detection in a large cohort of patients. Journal of the Neurological Sciences 193, 73–78. 10.1016/S0022-510X(01)00661-X [DOI] [PubMed] [Google Scholar]
  235. Swarup V, Phaneuf D, Dupré N, Petri S, Strong M, Kriz J, Julien J-P, 2011. Deregulation of TDP-43 in amyotrophic lateral sclerosis triggers nuclear factor κB–mediated pathogenic pathways. Journal of Experimental Medicine 208, 2429–2447. 10.1084/jem.20111313 [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Takano T, Kang J, Jaiswal JK, Simon SM, Lin JH-C, Yu Y, Li Y, Yang J, Dienel G, Zielke HR, Nedergaard M, 2005. Receptor-mediated glutamate release from volume sensitive channels in astrocytes. Proc Natl Acad Sci U S A 102, 16466–16471. 10.1073/pnas.0506382102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Takeuchi H, Kobayashi Y, Ishigaki S, Doyu M, Sobue G, 2002. Mitochondrial localization of mutant superoxide dismutase 1 triggers caspase-dependent cell death in a cellular model of familial amyotrophic lateral sclerosis. J. Biol. Chem 277, 50966–50972. 10.1074/jbc.M209356200 [DOI] [PubMed] [Google Scholar]
  238. Takuma H, Kwak S, Yoshizawa T, Kanazawa I, 1999. Reduction of GluR2 RNA editing, a molecular change that increases calcium influx through AMPA receptors, selective in the spinal ventral gray of patients with amyotrophic lateral sclerosis. Annals of Neurology 46, 806–815. [DOI] [PubMed] [Google Scholar]
  239. Tansey MG, Goldberg MS, 2010. Neuroinflammation in Parkinson’s disease: Its role in neuronal death and implications for therapeutic intervention. Neurobiology of Disease, Inflammation in Neuropsychiatric Disease 37, 510–518. 10.1016/j.nbd.2009.11.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  240. Taylor JP, Brown RH Jr, Cleveland DW, 2016. Decoding ALS: from genes to mechanism. Nature 539, 197–206. 10.1038/nature20413 [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Tcw J, Wang M, Pimenova AA, Bowles KR, Hartley BJ, Lacin E, Machlovi SI, Abdelaal R, Karch CM, Phatnani H, Slesinger PA, Zhang B, Goate AM, Brennand KJ, 2017. An Efficient Platform for Astrocyte Differentiation from Human Induced Pluripotent Stem Cells. Stem Cell Reports 9, 600–614. 10.1016/j.stemcr.2017.06.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Tian E, Sun Guoqiang, Sun Guihua, Chao J, Ye P, Warden C, Riggs AD, Shi Y, 2016. Small-Molecule-Based Lineage Reprogramming Creates Functional Astrocytes. Cell Reports 16, 781–792. 10.1016/j.celrep.2016.06.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  243. Tohidpour A, Morgun AV, Boitsova EB, Malinovskaya NA, Martynova GP, Khilazheva ED, Kopylevich NV, Gertsog GE, Salmina AB, 2017. Neuroinflammation and Infection: Molecular Mechanisms Associated with Dysfunction of Neurovascular Unit. Front. Cell. Infect. Microbiol 7 10.3389/fcimb.2017.00276 [DOI] [PMC free article] [PubMed] [Google Scholar]
  244. Tong J, Huang C, Bi F, Wu Q, Huang B, Liu X, Li F, Zhou H, Xia X-G, 2013. Expression of ALS-linked TDP-43 mutant in astrocytes causes non-cell-autonomous motor neuron death in rats. EMBO J 32, 1917–1926. 10.1038/emboj.2013.122 [DOI] [PMC free article] [PubMed] [Google Scholar]
  245. Topp JD, Gray NW, Gerard RD, Horazdovsky BF, 2004. Alsin Is a Rab5 and Rac1 Guanine Nucleotide Exchange Factor. J. Biol. Chem 279, 24612–24623. 10.1074/jbc.M313504200 [DOI] [PubMed] [Google Scholar]
  246. Tripathi P, Rodriguez-Muela N, Klim JR, de Boer AS, Agrawal S, Sandoe J, Lopes CS, Ogliari KS, Williams LA, Shear M, Rubin LL, Eggan K, Zhou Q, 2017. Reactive Astrocytes Promote ALS-like Degeneration and Intracellular Protein Aggregation in Human Motor Neurons by Disrupting Autophagy through TGF-β1. Stem Cell Reports 9, 667–680. 10.1016/j.stemcr.2017.06.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  247. Tsuda H, Han SM, Yang Y, Tong C, Lin YQ, Mohan K, Haueter C, Zoghbi A, Harati Y, Kwan J, Miller MA, Bellen HJ, 2008. The Amyotrophic Lateral Sclerosis 8 Protein VAPB Is Cleaved, Secreted, and Acts as a Ligand for Eph Receptors. Cell 133, 963–977. 10.1016/j.cell.2008.04.039 [DOI] [PMC free article] [PubMed] [Google Scholar]
  248. Turner Bradley J, Murray Simon S, Piccenna Loretta G, Lopes Elizabeth C, Kilpatrick Trevor J, Cheema Surindar S, 2004. Effect of p75 neurotrophin receptor antagonist on disease progression in transgenic amyotrophic lateral sclerosis mice. Journal of Neuroscience Research 78, 193–199. 10.1002/jnr.20256 [DOI] [PubMed] [Google Scholar]
  249. Tyzack GE, Hall CE, Sibley CR, Cymes T, Forostyak S, Carlino G, Meyer IF, Schiavo G, Zhang S-C, Gibbons GM, Newcombe J, Patani R, Lakatos A, 2017. A neuroprotective astrocyte state is induced by neuronal signal EphB1 but fails in ALS models. Nat Commun 8 10.1038/s41467-017-01283-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  250. Van Damme P, Bogaert E, Dewil M, Hersmus N, Kiraly D, Scheveneels W, Bockx I, Braeken D, Verpoorten N, Verhoeven K, Timmerman V, Herijgers P, Callewaert G, Carmeliet P, Van Den Bosch L, Robberecht W, 2007. Astrocytes regulate GluR2 expression in motor neurons and their vulnerability to excitotoxicity. Proc. Natl. Acad. Sci. U.S.A 104, 14825–14830. 10.1073/pnas.0705046104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  251. Van Damme P, Van den Bosch L, Van Houtte E, Callewaert G, Robberecht W, 2002. GluR2-Dependent Properties of AMPA Receptors Determine the Selective Vulnerability of Motor Neurons to Excitotoxicity. Journal of Neurophysiology 88, 1279–1287. 10.1152/jn.2002.88.3.1279 [DOI] [PubMed] [Google Scholar]
  252. Van Den Bosch L, Schwaller B, Vleminckx V, Meijers B, Stork S, Ruehlicke T, Van Houtte E, Klaassen H, Celio MR, Missiaen L, Robberecht W, Berchtold MW, 2002. Protective Effect of Parvalbumin on Excitotoxic Motor Neuron Death. Experimental Neurology 174, 150–161. 10.1006/exnr.2001.7858 [DOI] [PubMed] [Google Scholar]
  253. van Rheenen W, Shatunov A, Dekker AM, McLaughlin RL, Diekstra FP, Pulit SL, van der Spek RAA, Võsa U, de Jong S, Robinson MR, Yang J, Fogh I, van Doormaal PT, Tazelaar GHP, Koppers M, Blokhuis AM, Sproviero W, Jones AR, Kenna KP, van Eijk KR, Harschnitz O, Schellevis RD, Brands WJ, Medic J, Menelaou A, Vajda A, Ticozzi N, Lin K, Rogelj B, Vrabec K, Ravnik-Glavač M, Koritnik B, Zidar J, Leonardis L, Grošelj LD, Millecamps S, Salachas F, Meininger V, de Carvalho M, Pinto S, Mora JS, Rojas-García R, Polak M, Chandran S, Colville S, Swingler R, Morrison KE, Shaw PJ, Hardy J, Orrell RW, Pittman A, Sidle K, Fratta P, Malaspina A, Topp S, Petri S, Abdulla S, Drepper C, Sendtner M, Meyer T, Ophoff RA, Staats KA, Wiedau-Pazos M, Lomen-Hoerth C, Van Deerlin VM, Trojanowski JQ, Elman L, McCluskey L, Basak AN, Tunca C, Hamzeiy H, Parman Y, Meitinger T, Lichtner P, Radivojkov-Blagojevic M, Andres CR, Maurel C, Bensimon G, Landwehrmeyer B, Brice A, Payan CAM, Saker-Delye S, Dürr A, Wood NW, Tittmann L, Lieb W, Franke A, Rietschel M, Cichon S, Nöthen MM, Amouyel P, Tzourio C, Dartigues J-F, Uitterlinden AG, Rivadeneira F, Estrada K, Hofman A, Curtis C, Blauw HM, van der Kooi AJ, de Visser M, Goris A, Weber M, Shaw CE, Smith BN, Pansarasa O, Cereda C, Bo RD, Comi GP, D’Alfonso S, Bertolin C, Sorarù G, Mazzini L, Pensato V, Gellera C, Tiloca C, Ratti A, Calvo A, Moglia C, Brunetti M, Arcuti S, Capozzo R, Zecca C, Lunetta C, Penco S, Riva N, Padovani A, Filosto M, Muller B, Stuit RJ, Blair I, Zhang K, McCann EP, Fifita JA, Nicholson GA, Rowe DB, Pamphlett R, Kiernan MC, Grosskreutz J, Witte OW, Ringer T, Prell T, Stubendorff B, Kurth I, Hübner CA, Leigh PN, Casale F, Chio A, Beghi E, Pupillo E, Tortelli R, Logroscino G, Powell J, Ludolph AC, Weishaupt JH, Robberecht W, Van Damme P, Franke L, Pers TH, Brown RH, Glass JD, Landers JE, Hardiman O, Andersen PM, Corcia P, Vourc’h P, Silani V, Wray NR, Visscher PM, de Bakker PIW, van Es MA, Pasterkamp RJ, Lewis CM, Breen G, Al-Chalabi A, van den Berg LH, Veldink JH, 2016. Genome-wide association analyses identify new risk variants and the genetic architecture of amyotrophic lateral sclerosis. Nat Genet 48, 1043–1048. 10.1038/ng.3622 [DOI] [PMC free article] [PubMed] [Google Scholar]
  254. Vandenberghe W, Robberecht W, Brorson JR, 2000. AMPA Receptor Calcium Permeability, GluR2 Expression, and Selective Motoneuron Vulnerability. J. Neurosci 20, 123–132. 10.1523/JNEUROSCI.20-01-00123.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
  255. Varcianna A, Myszczynska MA, Castelli LM, O’Neill B, Kim Y, Talbot J, Nyberg S, Nyamali I, Heath PR, Stopford MJ, Hautbergue GM, Ferraiuolo L, 2019. Micro-RNAs secreted through astrocyte-derived extracellular vesicles cause neuronal network degeneration in C9orf72 ALS. EBioMedicine 40, 626–635. 10.1016/j.ebiom.2018.11.067 [DOI] [PMC free article] [PubMed] [Google Scholar]
  256. Vargas MR, Johnson DA, Sirkis DW, Messing A, Johnson JA, 2008a. Nrf2 Activation in Astrocytes Protects against Neurodegeneration in Mouse Models of Familial Amyotrophic Lateral Sclerosis. J. Neurosci 28, 13574–13581. 10.1523/JNEUROSCI.4099-08.2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  257. Vargas MR, Pehar M, Díaz-Amarilla PJ, Beckman JS, Barbeito L, 2008b. Transcriptional profile of primary astrocytes expressing ALS-linked mutant SOD1. J Neurosci Res 86, 3515–3525. 10.1002/jnr.21797 [DOI] [PMC free article] [PubMed] [Google Scholar]
  258. Velde CV, Miller TM, Cashman NR, Cleveland DW, 2008. Selective association of misfolded ALS-linked mutant SOD1 with the cytoplasmic face of mitochondria. PNAS 105, 4022–4027. 10.1073/pnas.0712209105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  259. Vucic S, Kiernan MC, 2006. Novel threshold tracking techniques suggest that cortical hyperexcitability is an early feature of motor neuron disease. Brain 129, 2436–2446. 10.1093/brain/awl172 [DOI] [PubMed] [Google Scholar]
  260. Vucic S, Nicholson GA, Kiernan MC, 2008. Cortical hyperexcitability may precede the onset of familial amyotrophic lateral sclerosis. Brain 131, 1540–1550. 10.1093/brain/awn071 [DOI] [PubMed] [Google Scholar]
  261. Wainger BJ, Kiskinis E, Mellin C, Wiskow O, Han SSW, Sandoe J, Perez NP, Williams LA, Lee S, Boulting G, Berry JD, Brown RH, Cudkowicz ME, Bean BP, Eggan K, Woolf CJ, 2014. Intrinsic Membrane Hyperexcitability of Amyotrophic Lateral Sclerosis Patient-Derived Motor Neurons. Cell Reports 7, 1–11. 10.1016/j.celrep.2014.03.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Waite AJ, Bäumer D, East S, Neal J, Morris HR, Ansorge O, Blake DJ, 2014. Reduced C9orf72 protein levels in frontal cortex of amyotrophic lateral sclerosis and frontotemporal degeneration brain with the C9ORF72 hexanucleotide repeat expansion. Neurobiol. Aging 35, 1779.e5–1779.e13. 10.1016/j.neurobiolaging.2014.01.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  263. Wall AA, Luo L, Hung Y, Tong SJ, Condon ND, Blumenthal A, Sweet MJ, Stow JL, 2017. Rab8a recruited PI3Kγ regulates signaling and cytokine outputs from endosomal Toll-like receptors. J. Biol. Chem jbc.M116.766337. 10.1074/jbc.M116.766337 [DOI] [PMC free article] [PubMed] [Google Scholar]
  264. Wang G, Anrather J, Huang J, Speth RC, Pickel VM, Iadecola C, 2004. NADPH oxidase contributes to angiotensin II signaling in the nucleus tractus solitarius. J. Neurosci 24, 5516–5524. 10.1523/JNEUROSCI.1176-04.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  265. Wang L, Gutmann DH, Roos RP, 2011. Astrocyte loss of mutant SOD1 delays ALS disease onset and progression in G85R transgenic mice. Hum Mol Genet 20, 286–293. 10.1093/hmg/ddq463 [DOI] [PubMed] [Google Scholar]
  266. Wang L, Yi F, Fu L, Yang J, Wang S, Wang Z, Suzuki K, Sun L, Xu X, Yu Y, Qiao J, Belmonte JCI, Yang Z, Yuan Y, Qu J, Liu G-H, 2017. CRISPR/Cas9-mediated targeted gene correction in amyotrophic lateral sclerosis patient iPSCs. Protein Cell 8, 365–378. 10.1007/s13238-017-0397-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  267. Wang Q, Liu Y, Zhou J, 2015. Neuroinflammation in Parkinson’s disease and its potential as therapeutic target. Transl Neurodegener 4 10.1186/s40035-015-0042-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  268. Watanabe S, Ilieva H, Tamada H, Nomura H, Komine O, Endo F, Jin S, Mancias P, Kiyama H, Yamanaka K, 2016. Mitochondria‐associated membrane collapse is a common pathomechanism in SIGMAR1‐ and SOD1‐linked ALS. EMBO Molecular Medicine 8, 1421–1437. 10.15252/emmm.201606403 [DOI] [PMC free article] [PubMed] [Google Scholar]
  269. Wen X, Tan W, Westergard T, Krishnamurthy K, Markandaiah SS, Shi Y, Lin S, Shneider NA, Monaghan J, Pandey UB, Pasinelli P, Ichida JK, Trotti D, 2014. Antisense Proline-Arginine RAN Dipeptides Linked to C9ORF72-ALS/FTD Form Toxic Nuclear Aggregates that Initiate In Vitro and In Vivo Neuronal Death. Neuron 84, 1213–1225. 10.1016/j.neuron.2014.12.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  270. West M, Mhatre M, Ceballos A, Floyd RA, Grammas P, Gabbita SP, Hamdheydari L, Mai T, Mou S, Pye QN, Stewart C, West S, Williamson KS, Zemlan F, Hensley K, 2004. The arachidonic acid 5-lipoxygenase inhibitor nordihydroguaiaretic acid inhibits tumor necrosis factor alpha activation of microglia and extends survival of G93A-SOD1 transgenic mice. J. Neurochem 91, 133–143. 10.1111/j.1471-4159.2004.02700.x [DOI] [PubMed] [Google Scholar]
  271. Westergard T, Jensen BK, Wen X, Cai J, Kropf E, Iacovitti L, Pasinelli P, Trotti D, 2016. Cell-to-Cell Transmission of Dipeptide Repeat Proteins Linked to C9orf72-ALS/FTD. Cell Reports 17, 645–652. 10.1016/j.celrep.2016.09.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  272. Williams KL, Topp S, Yang S, Smith B, Fifita JA, Warraich ST, Zhang KY, Farrawell N, Vance C, Hu X, Chesi A, Leblond CS, Lee A, Rayner SL, Sundaramoorthy V, Dobson-Stone C, Molloy MP, van Blitterswijk M, Dickson DW, Petersen RC, Graff-Radford NR, Boeve BF, Murray ME, Pottier C, Don E, Winnick C, McCann EP, Hogan A, Daoud H, Levert A, Dion PA, Mitsui J, Ishiura H, Takahashi Y, Goto J, Kost J, Gellera C, Gkazi AS, Miller J, Stockton J, Brooks WS, Boundy K, Polak M, Muñoz-Blanco JL, Esteban-Pérez J, Rábano A, Hardiman O, Morrison KE, Ticozzi N, Silani V, Belleroche J. de, Glass JD, Kwok JBJ, Guillemin GJ, Chung RS, Tsuji S, Jr, R.H.B., García-Redondo A, Rademakers R, Landers JE, Gitler AD, Rouleau GA, Cole NJ, Yerbury JJ, Atkin JD, Shaw CE, Nicholson GA, Blair IP, 2016. CCNF mutations in amyotrophic lateral sclerosis and frontotemporal dementia. Nature Communications 7, 11253 10.1038/ncomms11253 [DOI] [PMC free article] [PubMed] [Google Scholar]
  273. Wilms H, Sievers J, Rickert U, Rostami-Yazdi M, Mrowietz U, Lucius R, 2010. Dimethylfumarate inhibits microglial and astrocytic inflammation by suppressing the synthesis of nitric oxide, IL-1β, TNF-α and IL-6 in an in-vitro model of brain inflammation. Journal of Neuroinflammation 7, 30 10.1186/1742-2094-7-30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  274. Winkler EA, Sengillo JD, Sagare AP, Zhao Z, Ma Q, Zuniga E, Wang Y, Zhong Z, Sullivan JS, Griffin JH, Cleveland DW, Zlokovic BV, 2014. Blood–spinal cord barrier disruption contributes to early motor-neuron degeneration in ALS-model mice. PNAS 201401595. 10.1073/pnas.1401595111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  275. Wu D-C, Ré DB, Nagai M, Ischiropoulos H, Przedborski S, 2006. The inflammatory NADPH oxidase enzyme modulates motor neuron degeneration in amyotrophic lateral sclerosis mice. Proc Natl Acad Sci U S A 103, 12132–12137. 10.1073/pnas.0603670103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  276. Xu W-C, Liang J-Z, Li C, He Z-X, Yuan H-Y, Huang B-Y, Liu X-L, Tang B, Pang D-W, Du H-N, Yang Y, Chen J, Wang L, Zhang M, Liang Y, 2018. Pathological hydrogen peroxide triggers the fibrillization of wild-type SOD1 via sulfenic acid modification of Cys-111. Cell Death & Disease 9, 67 10.1038/s41419-017-0106-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  277. Xu Z, Yang C, 2014. TDP-43—The key to understanding amyotrophic lateral sclerosis. Rare Diseases 2, e944443 https://doi.org/10.416½1675511.2014.944443 [DOI] [PMC free article] [PubMed] [Google Scholar]
  278. Yamanaka K, Chun SJ, Boillee S, Fujimori-Tonou N, Yamashita H, Gutmann DH, Takahashi R, Misawa H, Cleveland DW, 2008. Astrocytes as determinants of disease progression in inherited ALS. Nat Neurosci 11, 251–253. 10.1038/nn2047 [DOI] [PMC free article] [PubMed] [Google Scholar]
  279. Yang Y, Hentati A, Deng H-X, Dabbagh O, Sasaki T, Hirano M, Hung W-Y, Ouahchi K, Yan J, Azim AC, Cole N, Gascon G, Yagmour A, Ben-Hamida M, Pericak-Vance M, Hentati F, Siddique T, 2001. The gene encoding alsin, a protein with three guanine-nucleotide exchange factor domains, is mutated in a form of recessive amyotrophic lateral sclerosis. Nature Genetics 29, 160–165. 10.1038/ng1001-160 [DOI] [PubMed] [Google Scholar]
  280. Ye Z-C, Wyeth MS, Baltan-Tekkok S, Ransom BR, 2003. Functional Hemichannels in Astrocytes: A Novel Mechanism of Glutamate Release. J. Neurosci 23, 3588–3596. 10.1523/JNEUROSCI.23-09-03588.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  281. Yi C, Mei X, Ezan P, Mato S, Matias I, Giaume C, Koulakoff A, 2016. Astroglial connexin43 contributes to neuronal suffering in a mouse model of Alzheimer’s disease. Cell Death Differ 23, 1691–1701. 10.1038/cdd.2016.63 [DOI] [PMC free article] [PubMed] [Google Scholar]
  282. Yoshihara T, Ishigaki S, Yamamoto M, Liang Y, Niwa J, Takeuchi H, Doyu M, Sobue G, 2002. Differential expression of inflammation- and apoptosis-related genes in spinal cords of a mutant SOD1 transgenic mouse model of familial amyotrophic lateral sclerosis. J. Neurochem 80, 158–167. [DOI] [PubMed] [Google Scholar]
  283. Yuva-Aydemir Y, Almeida S, Gao F-B, 2018. Insights into C9ORF72-Related ALS/FTD from Drosophila and iPSC Models. Trends Neurosci 41, 457–469. 10.1016/j.tins.2018.04.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  284. Zetterström P, Stewart HG, Bergemalm D, Jonsson PA, Graffmo KS, Andersen PM, Brännström T, Oliveberg M, Marklund SL, 2007. Soluble misfolded subfractions of mutant superoxide dismutase-1s are enriched in spinal cords throughout life in murine ALS models. Proc. Natl. Acad. Sci. U.S.A 104, 14157–14162. 10.1073/pnas.0700477104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  285. Zhan L, Xie Q, Tibbetts RS, 2015. Opposing roles of p38 and JNK in a Drosophila model of TDP-43 proteinopathy reveal oxidative stress and innate immunity as pathogenic components of neurodegeneration. Hum Mol Genet 24, 757–772. 10.1093/hmg/ddu493 [DOI] [PMC free article] [PubMed] [Google Scholar]
  286. Zhang F, Zhong R, Li S, Fu Z, Cheng C, Cai H, Le W, 2017. Acute Hypoxia Induced an Imbalanced M1/M2 Activation of Microglia through NF-κB Signaling in Alzheimer’s Disease Mice and Wild-Type Littermates. Front Aging Neurosci 9 10.3389/fnagi.2017.00282 [DOI] [PMC free article] [PubMed] [Google Scholar]
  287. Zhang K, Donnelly CJ, Haeusler AR, Grima JC, Machamer JB, Steinwald P, Daley EL, Miller SJ, Cunningham KM, Vidensky S, Gupta S, Thomas MA, Hong I, Chiu S-L, Huganir RL, Ostrow LW, Matunis MJ, Wang J, Sattler R, Lloyd TE, Rothstein JD, 2015. The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature 525, 56–61. 10.1038/nature14973 [DOI] [PMC free article] [PubMed] [Google Scholar]
  288. Zhang Q-J, Li J-J, Lin X, Lu Y-Q, Guo X-X, Dong E-L, Zhao M, He J, Wang N, Chen W-J, 2017. Modeling the phenotype of spinal muscular atrophy by the direct conversion of human fibroblasts to motor neurons. Oncotarget 8, 10945–10953. 10.18632/oncotarget.14641 [DOI] [PMC free article] [PubMed] [Google Scholar]
  289. Zhang Y, Chen K, Sloan SA, Bennett ML, Scholze AR, O’Keeffe S, Phatnani HP, Guarnieri P, Caneda C, Ruderisch N, Deng S, Liddelow SA, Zhang C, Daneman R, Maniatis T, Barres BA, Wu JQ, 2014. An RNA-Sequencing Transcriptome and Splicing Database of Glia, Neurons, and Vascular Cells of the Cerebral Cortex. J. Neurosci 34, 11929–11947. 10.1523/JNEUROSCI.1860-14.2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  290. Zhang Y, Sloan SA, Clarke LE, Caneda C, Plaza CA, Blumenthal PD, Vogel H, Steinberg GK, Edwards MSB, Li G, Duncan JA, Cheshier SH, Shuer LM, Chang EF, Grant GA, Gephart MGH, Barres BA, 2016. Purification and Characterization of Progenitor and Mature Human Astrocytes Reveals Transcriptional and Functional Differences with Mouse. Neuron 89, 37–53. 10.1016/j.neuron.2015.11.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  291. Zhang Y-J, Guo L, Gonzales PK, Gendron TF, Wu Y, Jansen-West K, O’Raw AD, Pickles SR, Prudencio M, Carlomagno Y, Gachechiladze MA, Ludwig C, Tian R, Chew J, DeTure M, Lin W-L, Tong J, Daughrity LM, Yue M, Song Y, Andersen JW, Castanedes-Casey M, Kurti A, Datta A, Antognetti G, McCampbell A, Rademakers R, Oskarsson B, Dickson DW, Kampmann M, Ward ME, Fryer JD, Link CD, Shorter J, Petrucelli L, 2019. Heterochromatin anomalies and double-stranded RNA accumulation underlie C9orf72 poly(PR) toxicity. Science 363, eaav2606. 10.1126/science.aav2606 [DOI] [PMC free article] [PubMed] [Google Scholar]
  292. Zhang Z, Almeida S, Lu Y, Nishimura AL, Peng L, Sun D, Wu B, Karydas AM, Tartaglia MC, Fong JC, Miller BL, Farese RV Jr, Moore MJ, Shaw CE, Gao F-B, 2013. Downregulation of MicroRNA-9 in iPSC-Derived Neurons of FTD/ALS Patients with TDP-43 Mutations. PLoS ONE 8, 1–11. 10.1371/journal.pone.0076055 [DOI] [PMC free article] [PubMed] [Google Scholar]
  293. Zimmerman MC, Sharma RV, Davisson RL, 2005. Superoxide mediates angiotensin II-induced influx of extracellular calcium in neural cells. Hypertension 45, 717–723. 10.1161/01.HYP.0000153463.22621.5e [DOI] [PubMed] [Google Scholar]
  294. Zufiría M, Gil-Bea FJ, Fernández-Torrón R, Poza JJ, Muñoz-Blanco JL, Rojas-García R, Riancho J, López de Munain A, 2016. ALS: A bucket of genes, environment, metabolism and unknown ingredients. Progress in Neurobiology 142, 104–129. 10.1016/j.pneurobio.2016.05.004 [DOI] [PubMed] [Google Scholar]

RESOURCES