Skip to main content
Erschienen in: Molecular Neurodegeneration 1/2017

Open Access 01.12.2017 | Review

Is amyotrophic lateral sclerosis/frontotemporal dementia an autophagy disease?

verfasst von: Zhiqiang Deng, Patricia Sheehan, Shi Chen, Zhenyu Yue

Erschienen in: Molecular Neurodegeneration | Ausgabe 1/2017

Abstract

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are neurodegenerative disorders that share genetic risk factors and pathological hallmarks. Intriguingly, these shared factors result in a high rate of comorbidity of these diseases in patients. Intracellular protein aggregates are a common pathological hallmark of both diseases. Emerging evidence suggests that impaired RNA processing and disrupted protein homeostasis are two major pathogenic pathways for these diseases. Indeed, recent evidence from genetic and cellular studies of the etiology and pathogenesis of ALS-FTD has suggested that defects in autophagy may underlie various aspects of these diseases. In this review, we discuss the link between genetic mutations, autophagy dysfunction, and the pathogenesis of ALS-FTD. Although dysfunction in a variety of cellular pathways can lead to these diseases, we provide evidence that ALS-FTD is, in many cases, an autophagy disease.
Abkürzungen
ALS
Amyotrophic lateral sclerosis
Atg
Autophagy-related genes
C9ORF72
Chromosome 9 open reading frame 72
CK2
Casein kinase 2
ER
Endoplasmic reticulum
FTD
Frontotemporal dementia
FTLD
Frontotemporal lobar degeneration
FUS
Fused in sarcoma
GEF
Guanine nucleotide exchange factor
GRD
Glycine-rich domain
HACE1
HECT domain and ankyrin repeat containing E3 Ub ligase 1
IBM
Inclusion body myopathy
IBs
Inclusion bodies
Keap1
Kelch-like ECH-associated protein 1
LC3
Microtubule-associated protein1 light chain 3
LIR
LC3 interacting regain
mTORC1
Mechanistic target of rapamycin complex 1
NBR1
Neighbor of BRCA1 gene 1
OPTN
Optineurin
PTPIP51
Protein tyrosine phosphatase interacting protein 51
RRM
RNA recognition motifs
SigR1
Sigma receptor-1
SMCR8
Smith-Magenis Chromosome Region gene 8
SOD1
Cu-Zn Superoxide Dismutase 1
TBK1
TANK-binding kinase 1
TDP-43
Transactive response DNA-binding protein 43
TFEB
Transcription factor EB
TRIM21
Tripartite motif-containing protein 21
UBA
ubiquitin-binding domain
UBQLN2
Ubiquilin 2
ULK1
Unc-51 Like Autophagy Activating Kinase 1
UPS
The ubiquitin-proteasome system
VAPB
Vesicle associated membrane protein associated protein B
VCP
Valosin containing protein
WDR41
WD repeat domain 41
WIPI2
WD-repeat protein interacting with phosphoinositides 2
YOD1
Yeast OTU deubiquinating enzyme 1

Background

Though various cellular defects are noted in Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal dementia (FTD) including dysregulation of RNA processing, protein aggregation, and oxidative stress, the detailed disease mechanisms remain poorly understood [1, 2]. Emerging evidence from genetic cases points to a role of autophagy in ALS-FTD. Here, we review how specific disease-linked mutations affect proper autophagic function and protein degradation leading to the speculation that ALS-FTD is, at least in part, an autophagy disease.

ALS and FTD

ALS is an adult-onset devastating neurodegenerative disorder characterized by progressive degeneration and deterioration of upper and lower motor neurons of the primary motor cortex, spinal cord and brain stem [3]. FTD is a form of dementia characterized by focal atrophy of the frontal and anterior temporal lobes of the brain, called frontaltemporal lobar degeneration (FTLD) [4]. The comorbidity of these diseases in patients and the shared genetic risk factors suggest ALS and FTD represent a continuum of neurodegenerative disorders [5]. A common pathological hallmark of both ALS and FTD is the presence of cytoplasmic protein aggregates and inclusions in affected neurons and glia cells [68], suggesting that an impairment in protein degradation may contribute to the disease process. In eukaryotic cells, the clearance of toxic-aggregated proteins is critical for cell survival, which relies mainly on two protein degradation systems: the ubiquitin-proteasome system (UPS) and autophagy [9, 10]. The UPS is mainly used for the degradation of short-lived proteins, while autophagy is preferentially used for the selective degradation of long-lived proteins and damaged organelles [11]. While the dysfunction of either the UPS or autophagy has been implicated in the formation of ALS-FTD-linked protein aggregates, accumulating evidence suggests that proper functioning of autophagy is the major determinant of motor neuron survival in ALS [1215].

Autophagic processing

Autophagy is a highly conserved catabolic cellular pathway used to degrade proteins and organelles at a basal level as well as pathogens and protein aggregates under pathological conditions. The endpoint of all autophagic pathways is the lysosome though, the three major autophagic pathways (i.e., microautophagy, chaperone-mediated autophagy, and macroautophagy) utilize different signaling molecules and protein machinery to prompt lysosomal degradation. This review will focus on the role of macroautophagy (hereafter referred to as autophagy) in ALS-FTD.
Canonical autophagy is under the control of mechanistic target of rapamycin complex 1 (mTORC1), a master regulator of diverse signaling pathways, which is regulated by cellular nutrient levels. Under nutrient-rich conditions, mTORC1 is active, suppressing the induction of autophagy by phosphorylating Unc51-like kinase 1 (ULK1) and Transcription Factor EB (TFEB). Starvation conditions inhibit the activity of mTORC1 and allow the induction of autophagy through the dephosphorylation of ULK1 and TFEB. Upon dephosphorylation, TFEB translocates to the nucleus where it functions to upregulate autophagy and lysosome-associated genes thus promoting autophagic degradation [16]. Once ULK1 is dephosphorylated, it forms a complex that can then phosphorylate Beclin1 [17] and Atg14L [18], two protein subunits in the class III phosphoinositol-3-phosphate kinase VPS34 complex. ULK1-mediated Atg14L phosphorylation activates VPS34 kinase activity, which is required for the nucleation of the phagophore membrane. The phagophore then expands to form the isolation membrane and engulf cytoplasmic contents. This elongation of the isolation membrane is dependent on two ubiquitin-like conjugation systems. The first involves the covalent conjugation of Atg12 to Atg5, a process carried out by Atg7 and Atg10. Atg5-Atg12 complex then binds Atg16. The second requires Atg7 and Atg3, assisted by Atg5-Atg12-Atg16, leading to the conjugation of phosphoethanolamine to LC3 [19]. This lipidation of LC3 allows it to associate with the autophagosome membrane and aid in cargo sequestration by associating with various autophagy receptors. Eventually, the membrane closes and forms a double membrane structure referred to as an autophagosome [19]. Following cargo sequestration, autophagosomes are trafficked along microtubule tracks. During this transport, autophagosomes can then fuse with compartments of the endocytic pathway to form an intermediate structure called an amphisome, which will then fuse with lysosomes for cargo degradation [20]. Additional studies investigating the cellular machinery for these fusion events have identified endosomal or lysosomal proteins (i.e., Rab7, LAMP1, HOPS complex proteins) and various SNARE proteins (i.e., Syntaxin17, SNAP29, VAMP7/8) as key mediators of autophagosome-lysosome fusion [21].
Autophagy was initially characterized as a non-selective degradation process. However, accumulating evidence shows that cargo such as damaged mitochondria and aggregated proteins can be selectively degraded by autophagy to maintain intracellular homeostasis [2224]. This process of selective autophagy is carried out by autophagy receptors, which recognize specific cargo (including disease-related proteins) through protein modifications of receptors in response to proteotoxic stress [25].
Although growing evidence points to an association of autophagic function and the development of ALS and FTD [8, 26], the precise role of autophagy in the pathogenesis of these diseases remains elusive. Here, we review recent evidence for the role of ALS-FTD-associated genes in autophagic processing.

ALS-FTD-associated genes and autophagy

C9ORF72

The GGGGCC (G4C2) hexanucleotide repeat expansion in C9ORF72 is the most common genetic cause of ALS-FTD [27]. Multiple pathogenic mechanisms are thought to underlie the pathology associated with the G4C2 expansion. This alteration of C9ORF72 may lead to epigenetic changes [28] that result in decreased mRNA levels and the loss-of-function of C9ORF72 [27]. Alternatively, G4C2 expansion containing transcripts may be subject to repeat-associated non-ATG (RAN) translation that could result in a toxic gain-of-function of dipeptide-repeat proteins [2931]. Multiple groups have initiated studies to develop therapeutic interventions aimed at reducing the toxicity of dipeptide-repeat proteins using gene depletion, peptide, or small molecule strategies to decrease RAN translation [32]. Pathologically, post-mortem analysis of patients carrying the G4C2 expansion in C9ORF72 reveals the presence of RNA foci and ubiquitin/p62 positive inclusions [3335]. Given these findings, much interest has arisen in understanding the potential link between C9ORF72 and autophagic regulation. Accumulating evidence demonstrates the relevance of C9ORF72 to autophagy, though its precise role in regulating autophagy is still unknown. Evidence suggests C9ORF72 acts in a multi-protein complex with SMCR8 and WDR41 to regulate the expression and activity of ULK1 [36]. Further, knockdown of C9ORF72 in MEF cells and mice impairs ULK1-mediated autophagy nduction, suggesting that C9ORF72 promotes autophagy [36, 37]. Acting in the same complex, C9ORF72 functions as a guanine nucleotide exchange factor (GEF) to activate Rab8a and Rab39b, two Rab GTPases with known functions in autophagy [38]. In the same study, depletion of C9ORF72 partly impaired autophagy and enhanced aggregation of TDP-43 and accumulation of p62 aggregates in neurons [38]. Additional studies indicate that C9ORF72 controls the Rab1a-dependent recruitment of the ULK1 complex to the phagophore in order to regulate the initiation of autophagy [39]. Again, the reduction of C9ORF72 protein levels was shown to attenuate autophagy and enhance the formation of p62-positive puncta in cells and primary neurons [39]. Together, these results indicate that C9ORF72 acts as a positive regulator of autophagy with its depletion leading to autophagic dysfunction and protein aggregation. However, in addition to regulating autophagy through direct association with the ULK1 autophagy initiation complex, C9ORF72 is indirectly involved in autophagy through regulation of mTORC1. Active mTORC1 inhibits autophagy by negatively regulating ULK1 activity and TFEB-mediated transcription of autophagy genes [4042]. In the context of ALS-FTD, depletion of C9ORF72 was shown to impair mTORC1 activity by decreasing the response of mTORC1 to amino acid availability [43], reducing the phosphorylation of S6 K, a substrate of mTORC1, and enhancing autophagic flux [44]. In contrast to the findings discussed earlier, these results suggest that depletion of C9ORF72 enhances autophagy through negative regulation of mTORC1. As it is still unclear what the precise role of C9ORF72 is in autophagy, further study is required to determine whether C9ORF72 depletion results in enhanced or impaired autophagy regulation.

TBK1

TANK-binding kinase 1 (TBK1) was recently identified as an ALS-FTD causal gene [45, 46]. ALS-FTD-linked TBK1 mutations are proposed to be loss-of-function mutations as patients show decreased mRNA and protein expression [45, 46]. TBK1 has been implicated in various autophagy-related pathways including antibacterial autophagy (xenophagy), mitophagy, and macroautophagy [4751]. TBK1 phosphorylates autophagy receptors including p62 and OPTN enhancing the elimination of intracellular pathogens such as Mycobacteria and Salmonella, as well as damaged mitochondria [4851]. Recent studies have shown that ALS-related mutations of TBK1 inhibit mitophagy by blocking autophagosome formation, disrupting the OPTN-TBK1 interaction [46, 52, 53], and failing to translocate to damaged mitochondria [54]. TBK1 is necessary for the maturation of the autophagosome, which is required for the degradation of p62 and its affiliated cargo [50]. Overexpression of wild-type TBK1, but not the kinase dead variant, facilitates clearance of mutant SOD1, another ALS associated protein [55, 56]. These results indicate that ALS-FTD-associated mutations of TBK1 disrupt autophagy through loss of TBK1-regulated clearance of autophagic cargo, including damaged mitochondria and disease-prone proteins.
Recently, a link between TBK1 and C9ORF72 has been reported. TBK1 phosphorylates SMCR8 at S402 and S796, which forms a complex with C9ORF72 and WDR41 to control autophagy flux [38]. Depletion of SMCR8 impairs autophagy, which can be rescued by expression of phosphomimetic variants of SMCR8 but not non-phosphorylatable variants of SMCR8, suggesting a critical role of TBK1 kinase activity in regulating autophagy through SMCR8. Moreover, knockdown of TBK1 enhances the accumulation of p62 puncta in neuronal cells, which can be rescued by phosphomimetic SMCR8 but not non-phosphorylatable SMCR8. These results imply that TBK1 and C9ORF72 may share a common pathway in regulation of autophagy through SMCR8 [38]. Together, ALS-FTD-causal mutations in TBK1 appear to cause the dysregulation of multiple aspects of autophagic function including mitophagy, toxic protein clearance, and C9ORF72/SMCR8 cellular pathways.

Optineurin (OPTN)

OPTN is involved in a variety of cellular functions, including signal transduction, vesicle trafficking, and axon homeostasis [57]. OPTN is a known autophagy receptor, which promotes the clearance of protein aggregates, infected pathogens, and mitochondria [48, 49, 55]. Several studies report that genetic mutations in OPTN enhance the risk of ALS [5863]. Multiple lines of evidence suggest that ALS related OPTN mutations impair autophagy. For example, ALS patients with OPTN mutation (E478G) show TDP-43 positive neuronal intracytoplasmic inclusions in medullary and spinal motor neurons [58, 59]. In addition, Wild et al. have shown that ALS-linked OPTN mutations (E478G and F178A) are unable to rescue antibacterial autophagy to the level of wild-type OPTN [48]. More recent studies have shown that ALS-linked mutations of OPTN (Q398X, E478G) disrupt its association with myosin VI, an association that is required for autophagosome maturation. This disruption leads to disturbances in the activation of ER stress, dysfunction in protein secretion, Golgi fragmentation, and autophagosome-lysosome fusion [64].
Post-translation modifications, including phosphorylation and ubiquitination, of OPTN enhance its function in autophagy. OPTN can be phosphorylated at various serine residues by TBK1. In antibacterial autophagy, TBK1 phosphorylates OPTN at S177, which enhances its binding affinity with LC3 and promotes autophagic clearance of cytosolic Salmonella [48]. While in mitophagy, TBK1 phosphorylates OPTN at S473 in the UBA domain, which enhances its binding affinity to ubiquitin chains [49]. In addition, phosphorylation of OPTN at Ser473, which is close to ALS-linked mutant site E478G, enhances its binding to ubiquitin chains and its clearance of damaged mitochondria [49]. However, interactome analysis in another study shows this mutation reduces OPTN interactions with binding partners, including proteins which functions in ubiquitin-dependent protein degradation and ER transport [65]. OPTN can also be ubiquitinated at multiple lysine residues by the tumor-suppressor HACE1, which promotes its interaction with p62 to form an autophagy receptor complex and accelerate autophagic flux [66]. Recently, Shen et al. have shown that compared with wild type OPTN, ubiquitin-binding domain mutants including the ALS-linked E478G and D474N mutations, are defective in clearing inclusion bodies formed by truncated TDP-43 [67]. The same group has also shown that the mutants act as dominant-negative traps to compromise the maturation of autophagosomes, which leads to defects in OPTN-mediated selective autophagy [67]. Future experiments are required to determine whether the phosphorylation or ubiquitination of OPTN is impacted in ALS linked mutations.
As discussed above here, post-translational modifications play a crucial role in modifying the function of OPTN in autophagy. Either phosphorylation or ubiquitination of OPTN increases autophagy and lead to the clearance of autophagy substrates. Therefore, it is conceivable that enhancing phosphorylation or ubiquitination of OPTN could be a novel therapeutic target for ALS patients carrying OPTN mutations.

p62 (SQSTM1)

p62 is an autophagy receptor that plays a major role in the clearance of protein aggregates [68]. Moreover, p62 itself is degraded by autophagy and thus can be used as a general marker for autophagic flux [68, 69]. p62 functions in diverse signaling pathways including proteasomal signaling, amino acid sensing, DNA damage response, and oxidative stress [63, 70, 71]. Pathological analyses of postmortem brains reveal that p62 associates with various disease-relevant protein aggregates [7274]. Due to the numerous roles of p62 in the clearance of pathogens, protein aggregates, and various client proteins, it has become the most well-characterized autophagy receptor [68, 69]. The UBA domain of p62 can be phosphorylated by Casein kinase 2 (CK2), TBK1, and ULK1. These phosphorylation events increase the affinity of p62 for ubiquitinated cargo and enhance the clearance of ubiquitinated proteins in selective autophagy [50, 68, 75]. Additionally, p62 can be ubiquitinated by TRIM21 and Keap1 to regulate the sequestration required for clearance of ubiquitinated proteins in selective autophagy [76, 77]. More recently, it has been reported that upon ubiquitin stress (e.g., proteasome inhibition), p62 undergoes ubiquitination which disrupts the dimerization at its UBA domain allowing p62 to recognize and bind polyubiquitinated cargoes [78].
Mutations in p62 have also been found in ALS-FTD patients [63, 7981]. A recent report indicates that some rare variants of p62 in the UBA domain increase FTLD risk [80]. While various mutations in p62 have been identified in ALS-FTD, the relevance of these mutations to disease risk is currently unknown [82]. Limited evidence suggests that disease-associated mutations may confer loss-of-function of p62 in selective autophagy [83, 84]. Knockdown of p62 in zebrafish causes a locomotor phenotype that is improved under mTOR inhibition or re-expression of wild-type p62, but not ALS-FTD-associated UBA domain mutant (P392L) p62 [83]. Further, the ALS-FTD-associated p62 LIR mutant, L341 V, is defective in recognition of LC3B [84]. This recognition is required for p62-mediated delivery of ubiquitinated cargo to the autophagosome suggesting this ALS-FTD related mutation results in failed selective autophagy. Finally, recent evidence suggests that p62 regulates the levels and/or functions of other ALS-FTD disease proteins (e.g., SOD1 and TDP-43) [85, 86], which is consistent with a prominent role of the misregulation of p62 in the disruption of protein homeostasis in ALS-FTD.

Ubiquilin 2 (Ubqln2)

Ubqln2 is ubiquitin-like receptor protein that shuttles polyubiquitinated cargo to the proteasome for degradation via its UBA domain [87]. While the role of Ubqln2 in the UPS has been well established, emerging evidence of an interaction between Ubqln2 and LC3 suggests this protein also has a role in autophagy [88, 89]. Multiple mutations in the PXX repeat region are linked to ALS-FTD [63, 90, 91]. In transgenic rats, neuronal expression of a disease-linked Ubqln2 mutation (P497H) induces the formation of protein aggregates positive for p62 and LC3, however, deletion of Ubqln2 has no effect [92]. As the effects of this mutation do not mimic loss-of-function, this finding suggests that the ALS-FTD-linked Ubqln2 mutation (P497H) confers a gain of toxic function [92]. Furthermore, expression of Ubqln2 mutants results in the accumulation of polyubiquitinated proteins (e.g., TDP-43) in a UBA domain-dependent manner [93]. However, a more recent study has shown that Ubqln2 clears aggregates via the proteasome by interacting with the chaperone protein HSP70 [94]. Disease mutations in Ubqln2 are defective in HSP70 binding leading to the accumulation of aggregated proteins, which suggests loss-of-function mutations in Ubqln2 may underlie ALS-FTD [94].

TDP-43

TDP-43 is an RNA- and DNA-binding protein, which harbors RNA recognition motifs (RRM) and a C-terminal glycine-rich domain (GRD) that are required for its functions in nucleic acid binding and protein-protein interactions, respectively [95]. TDP-43 was detected as a major component of the ubiquitin-positive inclusions in ALS-FTD [96]. During the past several years, the identification of different mutations in ALS-FTD has shown that TDP-43 is a causative gene to these diseases [97]. It is reported that TDP-43, especially aggregated TDP-43, is cleared by autophagy [85, 98, 99] and autophagy activators reduced the formation of TDP-43 positive inclusion in TDP-43 overexpression mouse models [100]. High levels of truncated TDP-43, called TDP-25, are a prominent feature of ALS-FTD and are associated with reduced induction of autophagy [101]. Others have reported that depletion of TDP-43 reduces the mRNA level of Atg7, by destabilizing Atg7 mRNA, which causes an impairment in autophagy and the accumulation of polyubiquitinated proteins and p62 in neuroblastoma cells [102]. However, a recent report showed that loss of TDP-43 increased the biogenesis of autophagosomes and lysosomes by inhibiting mTORC1 activity and enhancing TFEB activity while also disrupting the fusion of autophagosomes and lysosomes [103]. Although these studies show seemingly opposite effects of the loss of TDP-43 on autophagy induction, they each suggest an essential role of full-length TDP-43 in proper autophagy regulation and indicate the dysfunction of multiple steps of autophagy is linked to TDP-43-mediated ALS-FTD.
Stress granules are aggregations of RNA and RNA-binding proteins that are dynamically assembled and disassembled when cells are insulted by stress [104]. These stress granules are essential to the maintenance of RNA homeostasis, the dysregulation of which is highly implicated in ALS pathogenesis [104, 105]. TDP-43 is a major component of stress granules in ALS while another ALS-related protein, FUS, is found in these granules in a subset of cases [106, 107]. FUS is also an RNA binding protein and mutations in FUS account for around 5% of familial ALS cases and result in the loss-of-function of FUS [108, 109]. ALS-associated FUS mutation (R521C) accumulates FUS positive stress granules upon stress conditions and disrupts the release of FUS from stress granules in cultured neurons [110]. Moreover, ALS-linked mutant FUS affects the assembly and morphology of stress granules [111]. As stress granules are degraded by autophagy, it is conceivable that mutant FUS decreases the dynamics of stress granules and leads to their accumulation by disrupting autophagic clearance of these structures [105, 112]. One study suggested that prolonged stress granule formation might contribute to the pathogenesis of ALS. Interestingly, treatment of small molecule inhibitors of eIFα, which is directly correlated with stress granule formation, caused reduction of TDP-43 toxicity [113]. Decrease of TDP-43 toxicity by the same inhibitors has been also shown in other model systems [114], providing the support to the idea of targeting TDP-43 and stress granule in therapeutic development.

VCP

Valosin-contain protein (VCP), also called AAA-ATPase p97, functions in diverse signaling pathways, including endoplasmic reticulum (ER)-associated degradation, transcriptional regulation, DNA damage, and membrane dynamics [115118]. Loss-of-function mutations in VCP are linked to several human degenerative diseases, including inclusion body myopathy (IBM), ALS-FTD [119, 120]. Histopathological analysis of VCP-associated neurodegenerative diseases show that affected tissues contain prominent inclusions containing ubiquitin and TDP-43 [121123], and an accumulation of LC3 and p62 [124]. These findings, along with results from recent publications, suggest that VCP is involved in autophagy and specifically functions in the delivery of cargo to lysosomes for degradation [125]. Evidence from yeast has shown an essential role of the VCP orthologue, Cdc48, in the formation of autophagosomes [126] and multiple groups have reported the loss of VCP expression or activity leads to an accumulation of autophagosomes which fail to degrade aggregated proteins due to impaired autophagosome-lysosome fusion [104, 106]. Additional studies have found that mTORC is disrupted in VCP-related diseases, which leads to increased autophagosome biogenesis [127]. This finding, along with recent studies showing a critical role of VCP in the elimination of damaged lysosomes by autophagy, suggests the loss of VCP results in the overproduction of autophagosomes, a disruption in autolysosome formation, and the dysfunction of lysosomes [128]. Other studies have also shown a role for VCP in mitophagy and stress granule degradation [129], suggesting that VCP mutations result in ALS-FTD by disrupting multiple autophagic pathways.

SOD1

The Cu/Zn superoxide dismutase (SOD1) is an ALS causative gene in which mutations account for about 1% of sporadic and 10% of familial cases [130]. Mice expressing mutant SOD1 show an accumulation of aggregated proteins (including mutant SOD1) in the spinal cord, which is comparable with findings in human ALS [131]. Accumulating evidence demonstrates that genetic mutations of SOD1 induce ALS through a dominant toxic gain-of-function rather than the loss of enzymatic function [132, 133]. Previous reports have shown that autophagy can degrade mutant SOD1, preventing its toxicity [134] and antagonizing its cytoplasmic accumulation [135]. Recently, p62 and another ALS-associated gene, ALS2, have been shown to have additive protective roles in antagonizing the toxicity induced by mutant SOD1 [136]. In addition, it has been reported that inhibition of motor neuron autophagy in SOD1-G93A mice induces neuromuscular denervation in the early stages of the disease [137]. Another recent article has shown that bosutinib, which boosts autophagy, can improve the survival of ALS iPSC-derived motor neurons from patients with familial ALS caused by mutations in SOD1 [138]. It is likely that the autophagy-lysosome pathway is disrupted at asymptomatic stages in familial ALS-associated human SOD1-G93A as suggested in a transgenic model [139]. Additionally, the expression of TFEB and Beclin 1, two autophagy-related proteins, is altered in NSC-34 cells and in transgenic mice expressing SOD1-G93A mutation [140, 141]. In other studies, hyperactivity of the autophagy/lysosome pathway was detected in motor neurons of SOD1-linked ALS mice [142], which may account for the accumulation of autophagosomes in transgenic mice expressing mutant SOD1-G93A [143]. Taken together, these studies suggest that mutant SOD1 leads to a misregulation in autophagy. It is intriguing to propose that the disturbance of autophagy-lysosome pathway contributes to the toxicity induced by mutant SOD1, and modulation of autophagy may be a potential strategy for SOD1-mediated ALS treatment.

ALS2 (Alsin)

Mutations in ALS2 were identified in juvenile cases of motor neuron diseases, including ALS [144146]. Previous reports showed that ALS2 activates Rab5 to regulate the trafficking, fusion, and maturation of endosomes [147149]. The pathogenic mutations in ALS2 fail to activate Rab5 and disrupt the formation of amphisomes, a hybrid organelle formed through the fusion of autophagosomes and endosomes [150]. Loss of ALS2 in SOD1-H46H transgenic mice results in the accumulation of insoluble high molecular weight SOD1 but also ubiquitinated proteins and p62, while overexpression of ALS2 reduces the toxicity of mutant SOD1 in cultured motor neuronal cells [151]. More recently, ALS2, together with p62, was shown to play an additive protective role in antagonizing the toxicity of mutant SOD1 by promoting the clearance of insoluble mutant SOD1 [136]. These results suggest that the loss-of-function of ALS2, associated with impaired autophagy-induced protein clearance, leads to motor neuron disease associated pathogenesis.

VAPB

Vesicle-associated membrane protein-associated protein B (VAPB, also known as ALS8) is a member of highly conserved VAP family of ER transmembrane proteins [152]. VAPB plays a role in diverse functions including regulating calcium homeostasis in mitochondria and membrane trafficking in dendrites [153, 154]. More recently, Gomez-Suaga et al. have shown that VAPB and PTPIP51, a mitochondrial associated protein, regulate autophagy by modulating calcium delivery at ER-mitochondria contact sites [155]. Following the initial identification of a dominant missense mutation of VAPB in patients with a slowly progressive form of ALS [156], other mutations of VAPB have also been found to cause ALS [157, 158]. Knock-in mice expressing mutant VAPB-P56S show accumulation of cytoplasmic inclusion containing mutant VAPB and ubiquitinated proteins in motor neurons selectively, implicating that defects in autophagy contribute to the pathogenesis of VAPB-mediated ALS [159].

SigR1

Sigma receptor-1 (SigR1, also known as ALS16) is an ER chaperone protein involved in calcium signaling, ion channel activity, synaptic plasticity, and neuronal survival [160]. A mutation in SigR1 (E102Q) was identified as causative of juvenile ALS-FTD [161]. Emerging evidence has shown the association of SigR1 with autophagy. Vollrath et al. reported that knockdown of SigR1 in HEK 293 or NSC34 cells leads to impaired autophagic degradation. Additionally, in cells stably expressing autophagy reporter RFP-GFP-LC3, depletion of SigR1 disrupts the fusion of autophagosome to lysosome [162]. More recently, the SigR1-E102Q protein was found to form aggregates in ER, which alters the structure of the ER and further results in autophagy impairment by affecting the fusion of autophagosomes to lysosomes. Moreover, similar results were found in primary lymphoblastoid cells derived from familial ALS patients harboring the SigR1-E102Q mutation [163]. These studies imply that mutations in SigR1 result in the loss of its function in maintaining ER morphology and in autophagic processing.

Conclusions

While dysfunction in multiple cellular processes can underlie the development of ALS/FTD, an assortment of evidence demonstrates a role of autophagy dysfunction in these diseases. The above evidence suggests that different ALS/FTD linked mutations lead to dysfunction in all aspects of autophagy ranging from initiation and cargo recognition to autophagosome fusion with endosomes and lysosomes (Table 1 and Fig. 1). Dysfunction in any of these steps of autophagy leads to a disruption in protein homeostasis and results in the accumulation of toxic protein aggregates. While general enhancement of autophagy is likely to reduce these protein aggregates, the chronic upregulation of autophagy biogenesis can lead to an imbalance of autophagosome formation and clearance, resulting in neurotoxicity. More directed therapeutic interventions targeted at specific regulation of autophagy receptors or proteins are necessary to combat toxic protein accumulation effectively. For instance, specific compounds (e.g., HSF1 inhibitor) that manipulate p62 protein modifications can alter the ability of p62 to recognize and recruit autophagic cargo [164]. Additionally, targeting the activity of kinases involved in autophagy (e.g., ULK1 or TBK1, which modulate selective autophagy) could also provide benefits in protein quality control. However, these initiatives will require further studies in vivo and in vitro to determine how specific mutations alter autophagy and what potential avenues are available for therapeutic intervention.
Table 1
ALS-FTD genes and their disease mutations linked to autophagy
Gene symbol
Protein
Cellular functions
The effect of the genetic variants
Autophagy involvement
C9ORF72
C9orf72
Proteostasis and vesicle dynamics
Loss/gain of function
Regulates initiation of autophagy. [36, 37]
TBK1
TBK1 (TANK-binding kinase 1)
Proteostasis and immunity
Loss of function
Phosphorylates autophagy receptors (p62 and OPTN); regulates selective autophagy. [4851]
OPTN
Optineurin
Proteostasis, vesicle trafficking and axon homeostasis
Loss of function
A substrate of TBK1 and autophagy receptor protein; selective autophagy. [25, 48, 49]
SQSTM1
p62
Pproteostasis, amino acid sensing, DNA damage response, and oxidative stress
Loss of function
A substrate of TBK1 and ULK1; autophagy receptor; selective autophagy.[25, 50, 68]
UBQLN2
Ubiquilin-2 (UBQLN2)
Proteasome, proteostasis, and vesicle trafficking
Gain/loss of function
A potential autophagy receptor. [88, 89]
TARDBP
TDP-43
RNA regulation
Loss/gain of function
Regulates autophagy initiation and autophagosome-lysosome fusion. [103]
FUS
FUS (fused in sarcoma)
RNA regulation, and DNA damage repair
Loss of function
ALS-linked mutations, P525L and R522G, impair autophagy. [165]
VCP
Valosin-containing protein
ER-associated degradation, DNA damage, and membrane dynamics
Loss of function
Regulates the clearance of lysosomes. [128]
SOD1
Superoxide dismutase 1
Dismutation reaction
Gain of function
Mutant SOD1 disrupts autophagy. [139143]
ALS2
Alsin
Proteostasis and endosome biogenesis
Loss of function
Pathogenic mutations in ALS2 disrupt the formation of amphisomes. [150]
VAPB
Vesicle-associated membrane protein-associated protein B/C
Proteostasis, calcium homeostasis, and proteins trafficking
Loss of function
Regulates ER-mitochondrial contact. [155]
SigR1
Sigma receptor-1
Proteostasis, Ca2+ signaling, ion channel activity, synaptic plasticity.
Loss of function
Regulates autophagosome-lysosome fusion. [162, 163]

Acknowledgements

We are thankful to all members of Yue and Chen laboratories for the critical reading and discussion of the manuscript.

Funding

This work was partially supported by NIH grant R01 NS060123 (ZY); Young One Thousand TalentProgram of China (SC); China Scholarship Council Fellowship (ZD).

Availability of data and materials

Not applicable.
Not applicable.
All authors have given their consent for publication.

Competing interests

All authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Bozzo F, et al. Oxidative stress and mitochondrial damage in the pathogenesis of ALS: new perspectives. Neurosci Lett. 2017;636:3–8.PubMedCrossRef Bozzo F, et al. Oxidative stress and mitochondrial damage in the pathogenesis of ALS: new perspectives. Neurosci Lett. 2017;636:3–8.PubMedCrossRef
3.
Zurück zum Zitat Al-Chalabi A, Hardiman O. The epidemiology of ALS: a conspiracy of genes, environment and time. Nat Rev Neurol. 2013;9:617–28.PubMedCrossRef Al-Chalabi A, Hardiman O. The epidemiology of ALS: a conspiracy of genes, environment and time. Nat Rev Neurol. 2013;9:617–28.PubMedCrossRef
5.
Zurück zum Zitat van Langenhove T, et al. The molecular basis of the frontotemporal lobar degeneration–amyotrophic lateral sclerosis spectrum. Ann Med. 2012;44:817–28.PubMedPubMedCentralCrossRef van Langenhove T, et al. The molecular basis of the frontotemporal lobar degeneration–amyotrophic lateral sclerosis spectrum. Ann Med. 2012;44:817–28.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Arai T, et al. TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem Biophys Res Commun. 2006;351:602–11.PubMedCrossRef Arai T, et al. TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem Biophys Res Commun. 2006;351:602–11.PubMedCrossRef
8.
Zurück zum Zitat Gotzl JK, et al. Impaired protein degradation in FTLD and related disorders. Ageing Res Rev. 2016;32:122–39.PubMedCrossRef Gotzl JK, et al. Impaired protein degradation in FTLD and related disorders. Ageing Res Rev. 2016;32:122–39.PubMedCrossRef
11.
Zurück zum Zitat Rubinsztein DC. The roles of intracellular protein-degradation pathways in neurodegeneration. Nature. 2006;443:780–6.PubMedCrossRef Rubinsztein DC. The roles of intracellular protein-degradation pathways in neurodegeneration. Nature. 2006;443:780–6.PubMedCrossRef
12.
Zurück zum Zitat Ferrucci M, Fulceri F., Toti L, Soldani P, Siciliano G, Paparelli A, Fornai F. (2011) Protein clearing pathways in ALS. Arch Italiennesde Biol 149(1):121-149. Ferrucci M, Fulceri F., Toti L, Soldani P, Siciliano G, Paparelli A, Fornai F. (2011) Protein clearing pathways in ALS. Arch Italiennesde Biol 149(1):121-149.
13.
Zurück zum Zitat Pasquali L, Ruggieri S, Murri L, Paparelli A, Fornai F. Does autophagy worsen or improve the survival of dopaminergic neurons? Parkinsonism Relat Disord. 2009;15:S24–7.PubMedCrossRef Pasquali L, Ruggieri S, Murri L, Paparelli A, Fornai F. Does autophagy worsen or improve the survival of dopaminergic neurons? Parkinsonism Relat Disord. 2009;15:S24–7.PubMedCrossRef
14.
16.
Zurück zum Zitat Settembre C, et al. Signals from the lysosome: a control centre for cellular clearance and energy metabolism. Nat Rev Mol Cell Biol. 2013;14:283–96.PubMedPubMedCentralCrossRef Settembre C, et al. Signals from the lysosome: a control centre for cellular clearance and energy metabolism. Nat Rev Mol Cell Biol. 2013;14:283–96.PubMedPubMedCentralCrossRef
17.
18.
Zurück zum Zitat Wold MS, et al. ULK1-mediated phosphorylation of ATG14 promotes autophagy and is impaired in Huntington's disease models. Mol Neurodegener. 2016;11:76.PubMedPubMedCentralCrossRef Wold MS, et al. ULK1-mediated phosphorylation of ATG14 promotes autophagy and is impaired in Huntington's disease models. Mol Neurodegener. 2016;11:76.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Mizushima N, et al. The role of Atg proteins in autophagosome formation. Ann Rev Cell Dev Biol. 2011;27:107–32.CrossRef Mizushima N, et al. The role of Atg proteins in autophagosome formation. Ann Rev Cell Dev Biol. 2011;27:107–32.CrossRef
20.
Zurück zum Zitat Lamb CA, et al. The autophagosome: origins unknown, biogenesis complex. Nat Rev Mol Cell Biol. 2013;14:759–74.PubMedCrossRef Lamb CA, et al. The autophagosome: origins unknown, biogenesis complex. Nat Rev Mol Cell Biol. 2013;14:759–74.PubMedCrossRef
21.
Zurück zum Zitat Itakura E, et al. The hairpin-type tail-anchored SNARE syntaxin 17 targets to autophagosomes for fusion with endosomes/lysosomes. Cell. 2012;151:1256–69.PubMedCrossRef Itakura E, et al. The hairpin-type tail-anchored SNARE syntaxin 17 targets to autophagosomes for fusion with endosomes/lysosomes. Cell. 2012;151:1256–69.PubMedCrossRef
22.
Zurück zum Zitat Kraft C, et al. Selective types of autophagy in yeast. Biochim Biophys Acta. 2009;1793:1404–12.PubMedCrossRef Kraft C, et al. Selective types of autophagy in yeast. Biochim Biophys Acta. 2009;1793:1404–12.PubMedCrossRef
23.
Zurück zum Zitat Khaminets A, et al. Ubiquitin-dependent and independent signals in selective Autophagy. Trends Cell Biol. 2016;26:6–16.PubMedCrossRef Khaminets A, et al. Ubiquitin-dependent and independent signals in selective Autophagy. Trends Cell Biol. 2016;26:6–16.PubMedCrossRef
24.
Zurück zum Zitat Rogov V, et al. Interactions between autophagy receptors and ubiquitin-like proteins form the molecular basis for selective autophagy. Mol Cell. 2014;53:167–78.PubMedCrossRef Rogov V, et al. Interactions between autophagy receptors and ubiquitin-like proteins form the molecular basis for selective autophagy. Mol Cell. 2014;53:167–78.PubMedCrossRef
25.
Zurück zum Zitat Deng Z, et al. Autophagy receptors and neurodegenerative diseases. Trends Cell Biol. 2017;27:491–504.PubMedCrossRef Deng Z, et al. Autophagy receptors and neurodegenerative diseases. Trends Cell Biol. 2017;27:491–504.PubMedCrossRef
26.
Zurück zum Zitat Lee JK, et al. Role of autophagy in the pathogenesis of amyotrophic lateral sclerosis. Biochim Biophys Acta. 2015;1852:2517–24.PubMedCrossRef Lee JK, et al. Role of autophagy in the pathogenesis of amyotrophic lateral sclerosis. Biochim Biophys Acta. 2015;1852:2517–24.PubMedCrossRef
27.
Zurück zum Zitat DeJesus-Hernandez M, et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron. 2011;72:245–56.PubMedPubMedCentralCrossRef DeJesus-Hernandez M, et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron. 2011;72:245–56.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Belzil VV, et al. Reduced C9orf72 gene expression in c9FTD/ALS is caused by histone trimethylation, an epigenetic event detectable in blood. Acta Neuropathol. 2013;126:895–905.PubMedPubMedCentralCrossRef Belzil VV, et al. Reduced C9orf72 gene expression in c9FTD/ALS is caused by histone trimethylation, an epigenetic event detectable in blood. Acta Neuropathol. 2013;126:895–905.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Zhang YJ, et al. Aggregation-prone c9FTD/ALS poly(GA) RAN-translated proteins cause neurotoxicity by inducing ER stress. Acta Neuropathol. 2014;128:505–24.PubMedPubMedCentralCrossRef Zhang YJ, et al. Aggregation-prone c9FTD/ALS poly(GA) RAN-translated proteins cause neurotoxicity by inducing ER stress. Acta Neuropathol. 2014;128:505–24.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Lee YB, et al. C9orf72 poly GA RAN-translated protein plays a key role in amyotrophic lateral sclerosis via aggregation and toxicity. Hum Mol Genet. 2017;26(24):4765–77.PubMed Lee YB, et al. C9orf72 poly GA RAN-translated protein plays a key role in amyotrophic lateral sclerosis via aggregation and toxicity. Hum Mol Genet. 2017;26(24):4765–77.PubMed
32.
33.
Zurück zum Zitat Cooper-Knock J, et al. Clinico-pathological features in amyotrophic lateral sclerosis with expansions in C9ORF72. Brain J Neurol. 2012;135:751–64.CrossRef Cooper-Knock J, et al. Clinico-pathological features in amyotrophic lateral sclerosis with expansions in C9ORF72. Brain J Neurol. 2012;135:751–64.CrossRef
34.
Zurück zum Zitat Al-Sarraj S, et al. p62 positive, TDP-43 negative, neuronal cytoplasmic and intranuclear inclusions in the cerebellum and hippocampus define the pathology of C9orf72-linked FTLD and MND/ALS. Acta Neuropathol. 2011;122:691–702.PubMedCrossRef Al-Sarraj S, et al. p62 positive, TDP-43 negative, neuronal cytoplasmic and intranuclear inclusions in the cerebellum and hippocampus define the pathology of C9orf72-linked FTLD and MND/ALS. Acta Neuropathol. 2011;122:691–702.PubMedCrossRef
35.
Zurück zum Zitat Mackenzie IR, et al. The neuropathology associated with repeat expansions in the C9ORF72 gene. Acta Neuropathol. 2014;127:347–57.PubMedCrossRef Mackenzie IR, et al. The neuropathology associated with repeat expansions in the C9ORF72 gene. Acta Neuropathol. 2014;127:347–57.PubMedCrossRef
36.
Zurück zum Zitat Yang M, Liang C, Swaminathan K, et al. A C9ORF72 SMCR8-containing complex regulates ULK1 and plays a dual role in autophagy. Sci Adv. 2016;2(9):e1601167.PubMedPubMedCentralCrossRef Yang M, Liang C, Swaminathan K, et al. A C9ORF72 SMCR8-containing complex regulates ULK1 and plays a dual role in autophagy. Sci Adv. 2016;2(9):e1601167.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Sullivan PM, et al. The ALS/FTLD associated protein C9orf72 associates with SMCR8 and WDR41 to regulate the autophagy-lysosome pathway. Acta Neuropathol Commun. 2016;4:51.PubMedPubMedCentralCrossRef Sullivan PM, et al. The ALS/FTLD associated protein C9orf72 associates with SMCR8 and WDR41 to regulate the autophagy-lysosome pathway. Acta Neuropathol Commun. 2016;4:51.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Sellier C, et al. Loss of C9ORF72 impairs autophagy and synergizes with polyQ Ataxin-2 to induce motor neuron dysfunction and cell death. EMBO J. 2016;35:1276–97.PubMedPubMedCentralCrossRef Sellier C, et al. Loss of C9ORF72 impairs autophagy and synergizes with polyQ Ataxin-2 to induce motor neuron dysfunction and cell death. EMBO J. 2016;35:1276–97.PubMedPubMedCentralCrossRef
39.
41.
Zurück zum Zitat Hosokawa N, et al. Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex required for autophagy. Mol Biol Cell. 2009;20:1981–91.PubMedPubMedCentralCrossRef Hosokawa N, et al. Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex required for autophagy. Mol Biol Cell. 2009;20:1981–91.PubMedPubMedCentralCrossRef
44.
45.
Zurück zum Zitat Cirulli ET, Lasseigne BN, Petrovski S, Sapp PC, Dion PA, Leblond CS, Couthouis J, et al. Exome sequencing in amyotrophic lateral sclerosis identifies risk genes and pathways. Science. 2015;347:1436–41.PubMedPubMedCentralCrossRef Cirulli ET, Lasseigne BN, Petrovski S, Sapp PC, Dion PA, Leblond CS, Couthouis J, et al. Exome sequencing in amyotrophic lateral sclerosis identifies risk genes and pathways. Science. 2015;347:1436–41.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Freischmidt A, et al. Haploinsufficiency of TBK1 causes familial ALS and fronto-temporal dementia. Nat Neurosci. 2015;18:631–6.PubMedCrossRef Freischmidt A, et al. Haploinsufficiency of TBK1 causes familial ALS and fronto-temporal dementia. Nat Neurosci. 2015;18:631–6.PubMedCrossRef
49.
Zurück zum Zitat Richter B, et al. Phosphorylation of OPTN by TBK1 enhances its binding to Ub chains and promotes selective autophagy of damaged mitochondria. Proc Natl Acad Sci U S A. 2016;113:4039–44.PubMedPubMedCentralCrossRef Richter B, et al. Phosphorylation of OPTN by TBK1 enhances its binding to Ub chains and promotes selective autophagy of damaged mitochondria. Proc Natl Acad Sci U S A. 2016;113:4039–44.PubMedPubMedCentralCrossRef
50.
51.
Zurück zum Zitat Matsumoto G, et al. TBK1 controls autophagosomal engulfment of polyubiquitinated mitochondria through p62/SQSTM1 phosphorylation. Hum Mol Genet. 2015;24:4429–42.PubMedCrossRef Matsumoto G, et al. TBK1 controls autophagosomal engulfment of polyubiquitinated mitochondria through p62/SQSTM1 phosphorylation. Hum Mol Genet. 2015;24:4429–42.PubMedCrossRef
52.
Zurück zum Zitat Moore AS, Holzbaur EL. Dynamic recruitment and activation of ALS-associated TBK1 with its target optineurin are required for efficient mitophagy. Proc Natl Acad Sci U S A. 2016;113:E3349–58.PubMedPubMedCentralCrossRef Moore AS, Holzbaur EL. Dynamic recruitment and activation of ALS-associated TBK1 with its target optineurin are required for efficient mitophagy. Proc Natl Acad Sci U S A. 2016;113:E3349–58.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Li F, et al. Structural insights into the interaction and disease mechanism of neurodegenerative disease-associated optineurin and TBK1 proteins. Nat Commun. 2016;7:12708.PubMedPubMedCentralCrossRef Li F, et al. Structural insights into the interaction and disease mechanism of neurodegenerative disease-associated optineurin and TBK1 proteins. Nat Commun. 2016;7:12708.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Richter B, Sliter DA, Herhaus L, Stolz A, Wang C, Beli P, Zaffagnini G, Wild P, Martens S, Wagner SA, Youle RJ, Dikic I. Phosphorylation of OPTN by TBK1 enhances its binding to Ub chains and promotes selective autophagy of damaged mitochondria. PNAS. 2016;113:4039–44.PubMedPubMedCentralCrossRef Richter B, Sliter DA, Herhaus L, Stolz A, Wang C, Beli P, Zaffagnini G, Wild P, Martens S, Wagner SA, Youle RJ, Dikic I. Phosphorylation of OPTN by TBK1 enhances its binding to Ub chains and promotes selective autophagy of damaged mitochondria. PNAS. 2016;113:4039–44.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Korac J, et al. Ubiquitin-independent function of optineurin in autophagic clearance of protein aggregates. J Cell Sci. 2013;126:580–92.PubMedCrossRef Korac J, et al. Ubiquitin-independent function of optineurin in autophagic clearance of protein aggregates. J Cell Sci. 2013;126:580–92.PubMedCrossRef
56.
Zurück zum Zitat Gurney ME, Pu H, Chiu AY, Dal Canto MC, Polchow CY, Alexander DD, Caliendo J, Hentati A, Kwon YW, Deng H-X, Chen W, Zhai P, Sufit RL, Siddique T. Motor neuron degeneration in mice that express a human cu Zn superoxide dismutase mutation. Science. 1994;264:1772–5. Gurney ME, Pu H, Chiu AY, Dal Canto MC, Polchow CY, Alexander DD, Caliendo J, Hentati A, Kwon YW, Deng H-X, Chen W, Zhai P, Sufit RL, Siddique T. Motor neuron degeneration in mice that express a human cu Zn superoxide dismutase mutation. Science. 1994;264:1772–5.
58.
Zurück zum Zitat Maruyama H, Kawakami H. Optineurin and amyotrophic lateral sclerosis. Geriatr Gerontol Int. 2013;13:528–32.PubMedCrossRef Maruyama H, Kawakami H. Optineurin and amyotrophic lateral sclerosis. Geriatr Gerontol Int. 2013;13:528–32.PubMedCrossRef
59.
Zurück zum Zitat Kamada M, et al. Clinicopathologic features of autosomal recessive amyotrophic lateral sclerosis associated with optineurin mutation. Neuropathology. 2014;34:64–70.PubMedCrossRef Kamada M, et al. Clinicopathologic features of autosomal recessive amyotrophic lateral sclerosis associated with optineurin mutation. Neuropathology. 2014;34:64–70.PubMedCrossRef
60.
Zurück zum Zitat Kachaner D, et al. Toward an integrative view of Optineurin functions. Cell Cycle. 2012;11:2808–18.PubMedCrossRef Kachaner D, et al. Toward an integrative view of Optineurin functions. Cell Cycle. 2012;11:2808–18.PubMedCrossRef
61.
Zurück zum Zitat Li C, et al. Optineurin mutations in patients with sporadic amyotrophic lateral sclerosis in China. Amyotroph Lateral Sclerosis Frontotemporal Degeneration. 2015;16:485–9.CrossRef Li C, et al. Optineurin mutations in patients with sporadic amyotrophic lateral sclerosis in China. Amyotroph Lateral Sclerosis Frontotemporal Degeneration. 2015;16:485–9.CrossRef
62.
Zurück zum Zitat Pottier C, et al. Whole-genome sequencing reveals important role for TBK1 and OPTN mutations in frontotemporal lobar degeneration without motor neuron disease. Acta Neuropathol. 2015;130:77–92.PubMedPubMedCentralCrossRef Pottier C, et al. Whole-genome sequencing reveals important role for TBK1 and OPTN mutations in frontotemporal lobar degeneration without motor neuron disease. Acta Neuropathol. 2015;130:77–92.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Deng Z, et al. Autophagy receptors and neurodegenerative diseases. Trends Cell Bio. 2017;27:491–504. Deng Z, et al. Autophagy receptors and neurodegenerative diseases. Trends Cell Bio. 2017;27:491–504.
64.
Zurück zum Zitat Sundaramoorthy V, et al. Defects in optineurin- and myosin VI-mediated cellular trafficking in amyotrophic lateral sclerosis. Hum Mol Genet. 2015;24:3830–46.PubMedCrossRef Sundaramoorthy V, et al. Defects in optineurin- and myosin VI-mediated cellular trafficking in amyotrophic lateral sclerosis. Hum Mol Genet. 2015;24:3830–46.PubMedCrossRef
65.
Zurück zum Zitat Blokhuis AM, et al. Comparative interactomics analysis of different ALS-associated proteins identifies converging molecular pathways. Acta Neuropathol. 2016;132(2):175–96.PubMedPubMedCentralCrossRef Blokhuis AM, et al. Comparative interactomics analysis of different ALS-associated proteins identifies converging molecular pathways. Acta Neuropathol. 2016;132(2):175–96.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Liu Z, et al. Ubiquitylation of autophagy receptor Optineurin by HACE1 activates selective autophagy for tumor suppression. Cancer Cell. 2014;26:106–20.PubMedPubMedCentralCrossRef Liu Z, et al. Ubiquitylation of autophagy receptor Optineurin by HACE1 activates selective autophagy for tumor suppression. Cancer Cell. 2014;26:106–20.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Shen WC, et al. Mutations in the ubiquitin-binding domain of OPTN/optineurin interfere with autophagy-mediated degradation of misfolded proteins by a dominant-negative mechanism. Autophagy. 2015;11:685–700.PubMedPubMedCentralCrossRef Shen WC, et al. Mutations in the ubiquitin-binding domain of OPTN/optineurin interfere with autophagy-mediated degradation of misfolded proteins by a dominant-negative mechanism. Autophagy. 2015;11:685–700.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Lim J, et al. Proteotoxic stress induces phosphorylation of p62/SQSTM1 by ULK1 to regulate selective autophagic clearance of protein aggregates. PLoS Genet. 2015;11:e1004987.PubMedPubMedCentralCrossRef Lim J, et al. Proteotoxic stress induces phosphorylation of p62/SQSTM1 by ULK1 to regulate selective autophagic clearance of protein aggregates. PLoS Genet. 2015;11:e1004987.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Ohtsuka S, et al. SQSTM1/p62/A170 regulates the severity of Legionella pneumophila pneumonia by modulating inflammasome activity. Eur J Immunol. 2014;44:1084–92.PubMedCrossRef Ohtsuka S, et al. SQSTM1/p62/A170 regulates the severity of Legionella pneumophila pneumonia by modulating inflammasome activity. Eur J Immunol. 2014;44:1084–92.PubMedCrossRef
70.
72.
Zurück zum Zitat Mizuno Y, et al. Immunoreactivities of p62, an ubiqutin-binding protein, in the spinal anterior horn cells of patients with amyotrophic lateral sclerosis. J Neurol Sci. 2006;249:13–8.PubMedCrossRef Mizuno Y, et al. Immunoreactivities of p62, an ubiqutin-binding protein, in the spinal anterior horn cells of patients with amyotrophic lateral sclerosis. J Neurol Sci. 2006;249:13–8.PubMedCrossRef
73.
Zurück zum Zitat Arai T, et al. Neuronal and glial inclusions in frontotemporal dementia with or without motor neuron disease are immunopositive for p62. Neurosci Lett. 2003;342:41–4.PubMedCrossRef Arai T, et al. Neuronal and glial inclusions in frontotemporal dementia with or without motor neuron disease are immunopositive for p62. Neurosci Lett. 2003;342:41–4.PubMedCrossRef
74.
Zurück zum Zitat Hiji M, et al. White matter lesions in the brain with frontotemporal lobar degeneration with motor neuron disease: TDP-43-immunopositive inclusions co-localize with p62, but not ubiquitin. Acta Neuropathol. 2008;116:183–91.PubMedCrossRef Hiji M, et al. White matter lesions in the brain with frontotemporal lobar degeneration with motor neuron disease: TDP-43-immunopositive inclusions co-localize with p62, but not ubiquitin. Acta Neuropathol. 2008;116:183–91.PubMedCrossRef
75.
Zurück zum Zitat Matsumoto G, et al. Serine 403 phosphorylation of p62/SQSTM1 regulates selective autophagic clearance of ubiquitinated proteins. Mol Cell. 2011;44:279–89.PubMedCrossRef Matsumoto G, et al. Serine 403 phosphorylation of p62/SQSTM1 regulates selective autophagic clearance of ubiquitinated proteins. Mol Cell. 2011;44:279–89.PubMedCrossRef
76.
78.
Zurück zum Zitat Peng H, et al. Ubiquitylation of p62/sequestosome1 activates its autophagy receptor function and controls selective autophagy upon ubiquitin stress. Cell Res. 2017;27:657–74.PubMedCrossRef Peng H, et al. Ubiquitylation of p62/sequestosome1 activates its autophagy receptor function and controls selective autophagy upon ubiquitin stress. Cell Res. 2017;27:657–74.PubMedCrossRef
79.
Zurück zum Zitat Rea SL, et al. SQSTM1 mutations--bridging Paget disease of bone and ALS/FTLD. Exp Cell Res. 2014;325:27–37.PubMedCrossRef Rea SL, et al. SQSTM1 mutations--bridging Paget disease of bone and ALS/FTLD. Exp Cell Res. 2014;325:27–37.PubMedCrossRef
80.
81.
Zurück zum Zitat Rea SL, et al. New insights into the role of sequestosome 1/p62 mutant proteins in the pathogenesis of Paget's disease of bone. Endocr Rev. 2013;34:501–24.PubMedCrossRef Rea SL, et al. New insights into the role of sequestosome 1/p62 mutant proteins in the pathogenesis of Paget's disease of bone. Endocr Rev. 2013;34:501–24.PubMedCrossRef
82.
Zurück zum Zitat Cirulli ET, Lasseigne BN, Goldstein DB. Exome sequencing in amyotrophic lateral sclerosis identifies risk genes and pathways. Science. 2015;347:1436–41.PubMedPubMedCentralCrossRef Cirulli ET, Lasseigne BN, Goldstein DB. Exome sequencing in amyotrophic lateral sclerosis identifies risk genes and pathways. Science. 2015;347:1436–41.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Lattante S, et al. Sqstm1 knock-down causes a locomotor phenotype ameliorated by rapamycin in a zebrafish model of ALS/FTLD. Hum Mol Genet. 2015;24:1682–90.PubMedCrossRef Lattante S, et al. Sqstm1 knock-down causes a locomotor phenotype ameliorated by rapamycin in a zebrafish model of ALS/FTLD. Hum Mol Genet. 2015;24:1682–90.PubMedCrossRef
84.
Zurück zum Zitat Goode A, et al. Defective recognition of LC3B by mutant SQSTM1/p62 implicates impairment of autophagy as a pathogenic mechanism in ALS-FTLD. Autophagy. 2016;12:1–11.CrossRef Goode A, et al. Defective recognition of LC3B by mutant SQSTM1/p62 implicates impairment of autophagy as a pathogenic mechanism in ALS-FTLD. Autophagy. 2016;12:1–11.CrossRef
85.
Zurück zum Zitat Brady OA, et al. Regulation of TDP-43 aggregation by phosphorylation and p62/SQSTM1. J Neurochem. 2011;116:248–59.PubMedCrossRef Brady OA, et al. Regulation of TDP-43 aggregation by phosphorylation and p62/SQSTM1. J Neurochem. 2011;116:248–59.PubMedCrossRef
86.
Zurück zum Zitat Hadano S, et al. Functional links between SQSTM1 and ALS2 in the pathogenesis of ALS: cumulative impact on the protection against mutant SOD1-mediated motor dysfunction in mice. Hum Mol Genet. 2016;25(15):3321–40.PubMedCrossRef Hadano S, et al. Functional links between SQSTM1 and ALS2 in the pathogenesis of ALS: cumulative impact on the protection against mutant SOD1-mediated motor dysfunction in mice. Hum Mol Genet. 2016;25(15):3321–40.PubMedCrossRef
88.
89.
91.
Zurück zum Zitat Dillen L, et al. Explorative genetic study of UBQLN2 and PFN1 in an extended Flanders-Belgian cohort of frontotemporal lobar degeneration patients. Neurobiol Aging. 2013;34(1711):e1711–5. Dillen L, et al. Explorative genetic study of UBQLN2 and PFN1 in an extended Flanders-Belgian cohort of frontotemporal lobar degeneration patients. Neurobiol Aging. 2013;34(1711):e1711–5.
92.
Zurück zum Zitat Wu Q, et al. Pathogenic Ubqln2 gains toxic properties to induce neuron death. Acta Neuropathol. 2015;129:417–28.PubMedCrossRef Wu Q, et al. Pathogenic Ubqln2 gains toxic properties to induce neuron death. Acta Neuropathol. 2015;129:417–28.PubMedCrossRef
93.
Zurück zum Zitat Osaka M, et al. Disturbance of proteasomal and autophagic protein degradation pathways by amyotrophic lateral sclerosis-linked mutations in ubiquilin 2. Biochem Biophys Res Commun. 2016;472:324–31.PubMedCrossRef Osaka M, et al. Disturbance of proteasomal and autophagic protein degradation pathways by amyotrophic lateral sclerosis-linked mutations in ubiquilin 2. Biochem Biophys Res Commun. 2016;472:324–31.PubMedCrossRef
95.
Zurück zum Zitat Ayala YM, Zago P, Ambrogio AD’, Xu Y-F, Petrucelli L, Buratti E, Baralle FE. Structural determinants of the cellular localization and shuttling of TDP-43. J Cell Sci. 2008;121:3778–85.PubMedCrossRef Ayala YM, Zago P, Ambrogio AD’, Xu Y-F, Petrucelli L, Buratti E, Baralle FE. Structural determinants of the cellular localization and shuttling of TDP-43. J Cell Sci. 2008;121:3778–85.PubMedCrossRef
96.
Zurück zum Zitat Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, Bruce J, Schuck T, Grossman M, Clark CM, LF MC, Miller BL, Masliah E, Mackenzie IR, Feldman H, Feiden W, Kretzschmar HA, Trojanowski JQ, Lee VM. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314:130–3.PubMedCrossRef Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, Bruce J, Schuck T, Grossman M, Clark CM, LF MC, Miller BL, Masliah E, Mackenzie IR, Feldman H, Feiden W, Kretzschmar HA, Trojanowski JQ, Lee VM. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314:130–3.PubMedCrossRef
98.
Zurück zum Zitat Scotter EL, et al. Differential roles of the ubiquitin proteasome system and autophagy in the clearance of soluble and aggregated TDP-43 species. J Cell Sci. 2014;127:1263–78.PubMedPubMedCentralCrossRef Scotter EL, et al. Differential roles of the ubiquitin proteasome system and autophagy in the clearance of soluble and aggregated TDP-43 species. J Cell Sci. 2014;127:1263–78.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Wang X, et al. Degradation of TDP-43 and its pathogenic form by autophagy and the ubiquitin-proteasome system. Neurosci Lett. 2010;469:112–6.PubMedCrossRef Wang X, et al. Degradation of TDP-43 and its pathogenic form by autophagy and the ubiquitin-proteasome system. Neurosci Lett. 2010;469:112–6.PubMedCrossRef
100.
Zurück zum Zitat Wang I-F, G.B.-S., Liu Y-C, Wu C-C, Yang C-H, Tsai K-J, Shen C-KJ (2012) Autophagy activators rescue and alleviate pathogenesis of a mouse model with proteinopathies of the TAR DNA-binding protein 43. Proc Natl Acad Sci U S A 109: 15024 –15029. Wang I-F, G.B.-S., Liu Y-C, Wu C-C, Yang C-H, Tsai K-J, Shen C-KJ (2012) Autophagy activators rescue and alleviate pathogenesis of a mouse model with proteinopathies of the TAR DNA-binding protein 43. Proc Natl Acad Sci U S A 109: 15024 –15029.
101.
Zurück zum Zitat Caccamo A, et al. Reduced protein turnover mediates functional deficits in transgenic mice expressing the 25 kDa C-terminal fragment of TDP-43. Hum Mol Genet. 2015;24:4625–35.PubMedCrossRef Caccamo A, et al. Reduced protein turnover mediates functional deficits in transgenic mice expressing the 25 kDa C-terminal fragment of TDP-43. Hum Mol Genet. 2015;24:4625–35.PubMedCrossRef
103.
Zurück zum Zitat Xia Q, et al. TDP-43 loss of function increases TFEB activity and blocks autophagosome-lysosome fusion. EMBO J. 2016;35:121–42.PubMedCrossRef Xia Q, et al. TDP-43 loss of function increases TFEB activity and blocks autophagosome-lysosome fusion. EMBO J. 2016;35:121–42.PubMedCrossRef
106.
Zurück zum Zitat Bosco DA, et al. Mutant FUS proteins that cause amyotrophic lateral sclerosis incorporate into stress granules. Hum Mol Genet. 2010;19:4160–75.PubMedPubMedCentralCrossRef Bosco DA, et al. Mutant FUS proteins that cause amyotrophic lateral sclerosis incorporate into stress granules. Hum Mol Genet. 2010;19:4160–75.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Colombrita C, et al. TDP-43 is recruited to stress granules in conditions of oxidative insult. J Neurochem. 2009;111:1051–61.PubMedCrossRef Colombrita C, et al. TDP-43 is recruited to stress granules in conditions of oxidative insult. J Neurochem. 2009;111:1051–61.PubMedCrossRef
108.
109.
Zurück zum Zitat Kwiatkowski TJ, Bosco DA, Leclerc AL, Tamrazian E, Vanderburg CR, Russ C, et al. Mutations in the FUS TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science. 2009;323:1205–8.PubMedCrossRef Kwiatkowski TJ, Bosco DA, Leclerc AL, Tamrazian E, Vanderburg CR, Russ C, et al. Mutations in the FUS TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science. 2009;323:1205–8.PubMedCrossRef
110.
Zurück zum Zitat Ryu HH, et al. Autophagy regulates amyotrophic lateral sclerosis-linked fused in sarcoma-positive stress granules in neurons. Neurobiol Aging. 2014;35:2822–31.PubMedCrossRef Ryu HH, et al. Autophagy regulates amyotrophic lateral sclerosis-linked fused in sarcoma-positive stress granules in neurons. Neurobiol Aging. 2014;35:2822–31.PubMedCrossRef
111.
113.
Zurück zum Zitat Thomas MG, et al. RNA granules: the good, the bad and the ugly. Cell Signal. 2011;23:324–34.PubMedCrossRef Thomas MG, et al. RNA granules: the good, the bad and the ugly. Cell Signal. 2011;23:324–34.PubMedCrossRef
114.
Zurück zum Zitat Kim HJ, et al. Therapeutic modulation of eIF2alpha phosphorylation rescues TDP-43 toxicity in amyotrophic lateral sclerosis disease models. Nat Genet. 2014;46:152–60.PubMedCrossRef Kim HJ, et al. Therapeutic modulation of eIF2alpha phosphorylation rescues TDP-43 toxicity in amyotrophic lateral sclerosis disease models. Nat Genet. 2014;46:152–60.PubMedCrossRef
115.
Zurück zum Zitat Stolz A, et al. Cdc48: a power machine in protein degradation. Trends Biochem Sci. 2011;36:515–23.PubMedCrossRef Stolz A, et al. Cdc48: a power machine in protein degradation. Trends Biochem Sci. 2011;36:515–23.PubMedCrossRef
116.
Zurück zum Zitat Chapman E, et al. The complexities of p97 function in health and disease. Mol bio Syst. 2011;7:700–10. Chapman E, et al. The complexities of p97 function in health and disease. Mol bio Syst. 2011;7:700–10.
117.
Zurück zum Zitat Uchiyama K, Kondo H. p97/p47-mediated biogenesis of Golgi and ER. J Biochem. 2005;137:115–9.PubMedCrossRef Uchiyama K, Kondo H. p97/p47-mediated biogenesis of Golgi and ER. J Biochem. 2005;137:115–9.PubMedCrossRef
118.
Zurück zum Zitat Meyer H, et al. Emerging functions of the VCP/p97 AAA-ATPase in the ubiquitin system. Nat Cell Biol. 2012;14:117–23.PubMedCrossRef Meyer H, et al. Emerging functions of the VCP/p97 AAA-ATPase in the ubiquitin system. Nat Cell Biol. 2012;14:117–23.PubMedCrossRef
119.
Zurück zum Zitat Watts GD, et al. Inclusion body myopathy associated with Paget disease of bone and frontotemporal dementia is caused by mutant valosin-containing protein. Nat Genet. 2004;36:377–81.PubMedCrossRef Watts GD, et al. Inclusion body myopathy associated with Paget disease of bone and frontotemporal dementia is caused by mutant valosin-containing protein. Nat Genet. 2004;36:377–81.PubMedCrossRef
121.
Zurück zum Zitat Hubbers CU, et al. Pathological consequences of VCP mutations on human striated muscle. Brain J Neurol. 2007;130:381–93.CrossRef Hubbers CU, et al. Pathological consequences of VCP mutations on human striated muscle. Brain J Neurol. 2007;130:381–93.CrossRef
122.
Zurück zum Zitat Schroder R, et al. Mutant valosin-containing protein causes a novel type of frontotemporal dementia. Ann Neurol. 2005;57:457–61.PubMedCrossRef Schroder R, et al. Mutant valosin-containing protein causes a novel type of frontotemporal dementia. Ann Neurol. 2005;57:457–61.PubMedCrossRef
123.
Zurück zum Zitat Custer SK, et al. Transgenic mice expressing mutant forms VCP/p97 recapitulate the full spectrum of IBMPFD including degeneration in muscle, brain and bone. Hum Mol Genet. 2010;19:1741–55.PubMedCrossRef Custer SK, et al. Transgenic mice expressing mutant forms VCP/p97 recapitulate the full spectrum of IBMPFD including degeneration in muscle, brain and bone. Hum Mol Genet. 2010;19:1741–55.PubMedCrossRef
124.
125.
Zurück zum Zitat Dargemont C, Ossareh-Nazari B. Cdc48/p97, a key actor in the interplay between autophagy and ubiquitin/proteasome catabolic pathways. Biochim Biophys Acta. 2012;1823:138–44.PubMedCrossRef Dargemont C, Ossareh-Nazari B. Cdc48/p97, a key actor in the interplay between autophagy and ubiquitin/proteasome catabolic pathways. Biochim Biophys Acta. 2012;1823:138–44.PubMedCrossRef
126.
127.
Zurück zum Zitat Ching JK, et al. mTOR dysfunction contributes to vacuolar pathology and weakness in valosin-containing protein associated inclusion body myopathy. Hum Mol Genet. 2013;22:1167–79.PubMedCrossRef Ching JK, et al. mTOR dysfunction contributes to vacuolar pathology and weakness in valosin-containing protein associated inclusion body myopathy. Hum Mol Genet. 2013;22:1167–79.PubMedCrossRef
128.
Zurück zum Zitat Papadopoulos C, et al. VCP/p97 cooperates with YOD1, UBXD1 and PLAA to drive clearance of ruptured lysosomes by autophagy. EMBO J. 2017;36:135–50.PubMedCrossRef Papadopoulos C, et al. VCP/p97 cooperates with YOD1, UBXD1 and PLAA to drive clearance of ruptured lysosomes by autophagy. EMBO J. 2017;36:135–50.PubMedCrossRef
129.
131.
Zurück zum Zitat Kato S. Amyotrophic lateral sclerosis models and human neuropathology: similarities and differences. Acta Neuropathol. 2008;115:97–114.PubMedCrossRef Kato S. Amyotrophic lateral sclerosis models and human neuropathology: similarities and differences. Acta Neuropathol. 2008;115:97–114.PubMedCrossRef
132.
Zurück zum Zitat Galaleldeen A, et al. Structural and biophysical properties of metal-free pathogenic SOD1 mutants A4V and G93A. Arch Biochem Biophys. 2009;492:40–7.PubMedPubMedCentralCrossRef Galaleldeen A, et al. Structural and biophysical properties of metal-free pathogenic SOD1 mutants A4V and G93A. Arch Biochem Biophys. 2009;492:40–7.PubMedPubMedCentralCrossRef
133.
Zurück zum Zitat Schmidlin T, et al. Structural changes to monomeric CuZn superoxide dismutase caused by the familial amyotrophic lateral sclerosis-associated mutation A4V. Biophys J. 2009;97:1709–18.PubMedPubMedCentralCrossRef Schmidlin T, et al. Structural changes to monomeric CuZn superoxide dismutase caused by the familial amyotrophic lateral sclerosis-associated mutation A4V. Biophys J. 2009;97:1709–18.PubMedPubMedCentralCrossRef
134.
Zurück zum Zitat Kabuta T, et al. Degradation of amyotrophic lateral sclerosis-linked mutant cu,Zn-superoxide dismutase proteins by macroautophagy and the proteasome. J Biol Chem. 2006;281:30524–33.PubMedCrossRef Kabuta T, et al. Degradation of amyotrophic lateral sclerosis-linked mutant cu,Zn-superoxide dismutase proteins by macroautophagy and the proteasome. J Biol Chem. 2006;281:30524–33.PubMedCrossRef
135.
136.
Zurück zum Zitat Hadano S, et al. Functional links between SQSTM1 and ALS2 in the pathogenesis of ALS: cumulative impact on the protection against mutant SOD1-mediated motor dysfunction in mice. Hum Mol Genet. 2016;25:3321–40.PubMedCrossRef Hadano S, et al. Functional links between SQSTM1 and ALS2 in the pathogenesis of ALS: cumulative impact on the protection against mutant SOD1-mediated motor dysfunction in mice. Hum Mol Genet. 2016;25:3321–40.PubMedCrossRef
137.
Zurück zum Zitat Rudnick ND, Griffey CJ, Guarnieri P, Gerbino V, Wang X, Piersaint JA, Tapia JC, Rich MM, Maniatis T. Distinct roles for motor neuron autophagy early and late in the SOD1G93A mouse model of ALS. PNAS. 2017;114:E8294–303. Rudnick ND, Griffey CJ, Guarnieri P, Gerbino V, Wang X, Piersaint JA, Tapia JC, Rich MM, Maniatis T. Distinct roles for motor neuron autophagy early and late in the SOD1G93A mouse model of ALS. PNAS. 2017;114:E8294–303.
138.
Zurück zum Zitat Keiko Imamura YI, Inoue H. The Src c-Abl pathway is a potential therapeutic target in amyotrophic lateral sclerosis. Sci Transl Med. 2017;9:eaaf 3962.CrossRef Keiko Imamura YI, Inoue H. The Src c-Abl pathway is a potential therapeutic target in amyotrophic lateral sclerosis. Sci Transl Med. 2017;9:eaaf 3962.CrossRef
139.
140.
141.
Zurück zum Zitat Chen Y, Liu H, Guan Y, Wang Q, Zhou F, Jie L, Ju J, Pu L, Du H, Wang X. The altered autophagy mediated by TFEB in animal and cell models of amyotrophic lateral sclerosis. Am J Transl Res. 2015;7(9):1574–87.PubMedPubMedCentral Chen Y, Liu H, Guan Y, Wang Q, Zhou F, Jie L, Ju J, Pu L, Du H, Wang X. The altered autophagy mediated by TFEB in animal and cell models of amyotrophic lateral sclerosis. Am J Transl Res. 2015;7(9):1574–87.PubMedPubMedCentral
142.
143.
Zurück zum Zitat Morimoto N, et al. Increased autophagy in transgenic mice with a G93A mutant SOD1 gene. Brain Res. 2007;1167:112–7.PubMedCrossRef Morimoto N, et al. Increased autophagy in transgenic mice with a G93A mutant SOD1 gene. Brain Res. 2007;1167:112–7.PubMedCrossRef
144.
Zurück zum Zitat Hadano S, et al. Molecular and cellular function of ALS2/alsin: implication of membrane dynamics in neuronal development and degeneration. Neurochem Int. 2007;51:74–84.PubMedCrossRef Hadano S, et al. Molecular and cellular function of ALS2/alsin: implication of membrane dynamics in neuronal development and degeneration. Neurochem Int. 2007;51:74–84.PubMedCrossRef
145.
Zurück zum Zitat Eker HK, et al. A novel homozygous mutation in ALS2 gene in four siblings with infantile-onset ascending hereditary spastic paralysis. Eur J Med Genet. 2014;57:275–8.PubMedCrossRef Eker HK, et al. A novel homozygous mutation in ALS2 gene in four siblings with infantile-onset ascending hereditary spastic paralysis. Eur J Med Genet. 2014;57:275–8.PubMedCrossRef
146.
Zurück zum Zitat Wakil SM, et al. Infantile-onset ascending hereditary spastic paraplegia with bulbar involvement due to the novel ALS2 mutation c.2761C>T. Gene. 2014;536:217–20.PubMedCrossRef Wakil SM, et al. Infantile-onset ascending hereditary spastic paraplegia with bulbar involvement due to the novel ALS2 mutation c.2761C>T. Gene. 2014;536:217–20.PubMedCrossRef
147.
Zurück zum Zitat Topp JD, et al. Alsin is a Rab5 and Rac1 guanine nucleotide exchange factor. J Biol Chem. 2004;279:24612–23.PubMedCrossRef Topp JD, et al. Alsin is a Rab5 and Rac1 guanine nucleotide exchange factor. J Biol Chem. 2004;279:24612–23.PubMedCrossRef
148.
Zurück zum Zitat Kunita R, et al. The Rab5 activator ALS2/alsin acts as a novel Rac1 effector through Rac1-activated endocytosis. J Biol Chem. 2007;282:16599–611.PubMedCrossRef Kunita R, et al. The Rab5 activator ALS2/alsin acts as a novel Rac1 effector through Rac1-activated endocytosis. J Biol Chem. 2007;282:16599–611.PubMedCrossRef
149.
Zurück zum Zitat Otomo A. ALS2, a novel guanine nucleotide exchange factor for the small GTPase Rab5, is implicated in endosomal dynamics. Hum Mol Genet. 2003;12:1671–87.PubMedCrossRef Otomo A. ALS2, a novel guanine nucleotide exchange factor for the small GTPase Rab5, is implicated in endosomal dynamics. Hum Mol Genet. 2003;12:1671–87.PubMedCrossRef
150.
Zurück zum Zitat Otomo A, et al. Defective relocalization of ALS2/alsin missense mutants to Rac1-induced macropinosomes accounts for loss of their cellular function and leads to disturbed amphisome formation. FEBS Lett. 2011;585:730–6.PubMedCrossRef Otomo A, et al. Defective relocalization of ALS2/alsin missense mutants to Rac1-induced macropinosomes accounts for loss of their cellular function and leads to disturbed amphisome formation. FEBS Lett. 2011;585:730–6.PubMedCrossRef
151.
Zurück zum Zitat Kanekura K, et al. Alsin, the product of ALS2 gene, suppresses SOD1 mutant neurotoxicity through RhoGEF domain by interacting with SOD1 mutants. J Biol Chem. 2004;279:19247–56.PubMedCrossRef Kanekura K, et al. Alsin, the product of ALS2 gene, suppresses SOD1 mutant neurotoxicity through RhoGEF domain by interacting with SOD1 mutants. J Biol Chem. 2004;279:19247–56.PubMedCrossRef
152.
Zurück zum Zitat Lev S, et al. The VAP protein family: from cellular functions to motor neuron disease. Trends Cell Biol. 2008;18:282–90.PubMedCrossRef Lev S, et al. The VAP protein family: from cellular functions to motor neuron disease. Trends Cell Biol. 2008;18:282–90.PubMedCrossRef
153.
Zurück zum Zitat Kuijpers M, et al. The ALS8 protein VAPB interacts with the ER-Golgi recycling protein YIF1A and regulates membrane delivery into dendrites. EMBO J. 2013;32:2056–72.PubMedPubMedCentralCrossRef Kuijpers M, et al. The ALS8 protein VAPB interacts with the ER-Golgi recycling protein YIF1A and regulates membrane delivery into dendrites. EMBO J. 2013;32:2056–72.PubMedPubMedCentralCrossRef
154.
Zurück zum Zitat De Vos KJ, et al. VAPB interacts with the mitochondrial protein PTPIP51 to regulate calcium homeostasis. Hum Mol Genet. 2012;21:1299–311.PubMedCrossRef De Vos KJ, et al. VAPB interacts with the mitochondrial protein PTPIP51 to regulate calcium homeostasis. Hum Mol Genet. 2012;21:1299–311.PubMedCrossRef
156.
Zurück zum Zitat Nishimura, A.L. (2004) A novel locus for late onset amyotrophic lateral sclerosis/motor neurone disease variant at 20q13. J Med Genet 41, 315-320. Nishimura, A.L. (2004) A novel locus for late onset amyotrophic lateral sclerosis/motor neurone disease variant at 20q13. J Med Genet 41, 315-320.
157.
Zurück zum Zitat Kabashi E, et al. Investigating the contribution of VAPB/ALS8 loss of function in amyotrophic lateral sclerosis. Hum Mol Genet. 2013;22:2350–60.PubMedCrossRef Kabashi E, et al. Investigating the contribution of VAPB/ALS8 loss of function in amyotrophic lateral sclerosis. Hum Mol Genet. 2013;22:2350–60.PubMedCrossRef
158.
Zurück zum Zitat Chattopadhyay D, Sengupta S. First evidence of pathogenicity of V234I mutation of hVAPB found in amyotrophic lateral sclerosis. Biochem Biophys Res Commun. 2014;448:108–13.PubMedCrossRef Chattopadhyay D, Sengupta S. First evidence of pathogenicity of V234I mutation of hVAPB found in amyotrophic lateral sclerosis. Biochem Biophys Res Commun. 2014;448:108–13.PubMedCrossRef
159.
Zurück zum Zitat Larroquette F, et al. Vapb/amyotrophic lateral sclerosis 8 knock-in mice display slowly progressive motor behavior defects accompanying ER stress and autophagic response. Hum Mol Genet. 2015;24:6515–29.PubMedPubMedCentralCrossRef Larroquette F, et al. Vapb/amyotrophic lateral sclerosis 8 knock-in mice display slowly progressive motor behavior defects accompanying ER stress and autophagic response. Hum Mol Genet. 2015;24:6515–29.PubMedPubMedCentralCrossRef
160.
Zurück zum Zitat Hayashi T, Su TP. Sigma-1 receptor chaperones at the ER-mitochondrion interface regulate ca(2+) signaling and cell survival. Cell. 2007;131:596–610.PubMedCrossRef Hayashi T, Su TP. Sigma-1 receptor chaperones at the ER-mitochondrion interface regulate ca(2+) signaling and cell survival. Cell. 2007;131:596–610.PubMedCrossRef
161.
Zurück zum Zitat Al-Saif A, et al. A mutation in sigma-1 receptor causes juvenile amyotrophic lateral sclerosis. Ann Neurol. 2011;70:913–9.PubMedCrossRef Al-Saif A, et al. A mutation in sigma-1 receptor causes juvenile amyotrophic lateral sclerosis. Ann Neurol. 2011;70:913–9.PubMedCrossRef
162.
Zurück zum Zitat Vollrath JT, et al. Loss of function of the ALS protein SigR1 leads to ER pathology associated with defective autophagy and lipid raft disturbances. Cell Death Dis. 2014;5:e1290.PubMedPubMedCentralCrossRef Vollrath JT, et al. Loss of function of the ALS protein SigR1 leads to ER pathology associated with defective autophagy and lipid raft disturbances. Cell Death Dis. 2014;5:e1290.PubMedPubMedCentralCrossRef
163.
Zurück zum Zitat Dreser A, et al. The ALS-linked E102Q mutation in sigma receptor-1 leads to ER stress-mediated defects in protein homeostasis and dysregulation of RNA-binding proteins. Cell Death Differ. 2017;24:1655–71.PubMedPubMedCentralCrossRef Dreser A, et al. The ALS-linked E102Q mutation in sigma receptor-1 leads to ER stress-mediated defects in protein homeostasis and dysregulation of RNA-binding proteins. Cell Death Differ. 2017;24:1655–71.PubMedPubMedCentralCrossRef
164.
Zurück zum Zitat Watanabe Y, et al. HSF1 stress response pathway regulates autophagy receptor SQSTM1/p62-associated proteostasis. Autophagy. 2017;13:133–48.PubMedCrossRef Watanabe Y, et al. HSF1 stress response pathway regulates autophagy receptor SQSTM1/p62-associated proteostasis. Autophagy. 2017;13:133–48.PubMedCrossRef
165.
Metadaten
Titel
Is amyotrophic lateral sclerosis/frontotemporal dementia an autophagy disease?
verfasst von
Zhiqiang Deng
Patricia Sheehan
Shi Chen
Zhenyu Yue
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Molecular Neurodegeneration / Ausgabe 1/2017
Elektronische ISSN: 1750-1326
DOI
https://doi.org/10.1186/s13024-017-0232-6

Weitere Artikel der Ausgabe 1/2017

Molecular Neurodegeneration 1/2017 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.