Review Special Issues

Mislocalization, aggregation formation and defect in proteolysis in ALS

  • Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by motoneuron degeneration. The features observed in ALS are the mislocalization and aggregation of proteins in the affected motoneurons. The mutants of Cu/Zn superoxide dismutase (SOD1) or TAR DNA binding protein (TDP-43) that cause ALS have been reported to localize aberrantly. These aggregations contain the products of ALS causative genes, including SOD1 or TDP-43. Notably, TDP-43-positive aggregations have been identified even in sporadic ALS cases, indicating the importance of aggregate formation in the pathogenesis of ALS. Various proteins other than ALS causative gene products are also included in these aggregates. It is thought that the genetic mutation-induced conformational changes of proteins cause the aberrant redistribution and formation of aggregates, resulting in a loss of function or a gain of neuronal toxicity through the undesired interactions. Additionally, valosin-containing protein (VCP), ubiquilin2 (UBQLN2) and optineurin (OPTN), which are related to the proteolysis system, have also been identified as causative genes in ALS. These facts suggest that the aberrant protein homeostasis mediated by mislocalization, aggregate formation, or defects in the proteolysis system are the underlying causes of neuronal toxicity in ALS. Here, we focus on the impaired protein homeostasis observed in ALS to discuss the potential for motoneuron toxicity.

    Citation: Atsushi Tsubota, Hidenori Ichijo, Kengo Homma. Mislocalization, aggregation formation and defect in proteolysis in ALS[J]. AIMS Molecular Science, 2016, 3(2): 246-268. doi: 10.3934/molsci.2016.2.246

    Related Papers:

    [1] Giulia Ambrosi, Pamela Milani . Endoplasmic reticulum, oxidative stress and their complex crosstalk in neurodegeneration: proteostasis, signaling pathways and molecular chaperones. AIMS Molecular Science, 2017, 4(4): 424-444. doi: 10.3934/molsci.2017.4.424
    [2] Marco Feligioni, Serena Marcelli, Erin Knock, Urooba Nadeem, Ottavio Arancio, Paul E. Fraser . SUMO modulation of protein aggregation and degradation. AIMS Molecular Science, 2015, 2(4): 382-410. doi: 10.3934/molsci.2015.4.382
    [3] Ashok Chakraborty, Anil Diwan . Molecular mechanisms of neurodegenerative disease (NDD). AIMS Molecular Science, 2023, 10(3): 171-185. doi: 10.3934/molsci.2023012
    [4] Isabel Saez, David Vilchez . Protein clearance mechanisms and their demise in age-related neurodegenerative diseases. AIMS Molecular Science, 2015, 1(1): 1-21. doi: 10.3934/molsci.2015.1.1
    [5] Ann De Vos, Tine Bynens, Joëlle Rosseels, Catherina Coun, Julia Ring, Frank Madeo, Marie-Christine Galas, Joris Winderickx, Vanessa Franssens . The peptidyl prolyl cis/trans isomerase Pin1/Ess1 inhibits phosphorylation and toxicity of tau in a yeast model for Alzheimer's disease. AIMS Molecular Science, 2015, 2(2): 144-160. doi: 10.3934/molsci.2015.2.144
    [6] Leena Latonen . Protein aggregation in neurodegenerative disease: the nucleolar connection. AIMS Molecular Science, 2015, 2(3): 324-331. doi: 10.3934/molsci.2015.3.324
    [7] Rosanna Parlato, Holger Bierhoff . Role of nucleolar dysfunction in neurodegenerative disorders: a game of genes?. AIMS Molecular Science, 2015, 2(3): 211-224. doi: 10.3934/molsci.2015.3.211
    [8] Naomi L. Pollock, Oscar Moran, Debora Baroni, Olga Zegarra-Moran, Robert C. Ford . Characterisation of the salmon cystic fibrosis transmembrane conductance regulator protein for structural studies. AIMS Molecular Science, 2014, 1(4): 141-161. doi: 10.3934/molsci.2014.4.141
    [9] William M. Scovell, Sachindra R. Joshi . The changing paradigm: estrogen receptor α recognition on DNA and within the dynamic nature of nucleosomes. AIMS Molecular Science, 2015, 2(2): 48-63. doi: 10.3934/molsci.2015.2.48
    [10] Dora Brites . Cell ageing: a flourishing field for neurodegenerative diseases. AIMS Molecular Science, 2015, 2(3): 225-258. doi: 10.3934/molsci.2015.3.225
  • Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by motoneuron degeneration. The features observed in ALS are the mislocalization and aggregation of proteins in the affected motoneurons. The mutants of Cu/Zn superoxide dismutase (SOD1) or TAR DNA binding protein (TDP-43) that cause ALS have been reported to localize aberrantly. These aggregations contain the products of ALS causative genes, including SOD1 or TDP-43. Notably, TDP-43-positive aggregations have been identified even in sporadic ALS cases, indicating the importance of aggregate formation in the pathogenesis of ALS. Various proteins other than ALS causative gene products are also included in these aggregates. It is thought that the genetic mutation-induced conformational changes of proteins cause the aberrant redistribution and formation of aggregates, resulting in a loss of function or a gain of neuronal toxicity through the undesired interactions. Additionally, valosin-containing protein (VCP), ubiquilin2 (UBQLN2) and optineurin (OPTN), which are related to the proteolysis system, have also been identified as causative genes in ALS. These facts suggest that the aberrant protein homeostasis mediated by mislocalization, aggregate formation, or defects in the proteolysis system are the underlying causes of neuronal toxicity in ALS. Here, we focus on the impaired protein homeostasis observed in ALS to discuss the potential for motoneuron toxicity.


    1. Introduction

    Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by selective motoneuron cell death. Quadriplegia and respiratory disorders can occur during the disease progression, leading to death usually within 3-5 years after the onset of the disease. ALS is classified as either familial or sporadic, with familial ALS accounting for approximately 10% of all ALS cases. In 1993, Cu/Zn superoxide dismutase(SOD1) was first identified as a causal gene of familial ALS [1]. Since the discovery of TAR DNA binding protein(TARDBP) in 2008, which encodes TDP-43, another ALS causative gene [2], approximately 20 additional ALS causative genes have been reported due to the development of genetic techniques, such as the next generation sequencer. Notably, the distinguishing GC repeat expansion in the intronic region of the Chromosome 9 open reading frame 72 (C9ORF72) occurs in approximately 40% of all familial ALS cases [3,4,5]. In spite of these remarkable advances in inherited causal genes, the pathogenic mechanisms of familial and sporadic ALS still remain unclear. Therefore, there is a need to reveal the common motoneuron toxicity in ALS.

    One of the hallmarks of ALS is the dysregulation of protein localization. The formation of aggregates was predicated to be involved in the pathogenesis of ALS. Several causative gene mutations, including SOD1, TARDBP, and fused in sarcoma(FUS), have been reported to be mislocalized or form abnormal protein aggregates [1,6,7,8]. The mislocalization and aggregation of TDP-43 and FUS are observed even in sporadic ALS cases, suggesting that a defect in protein homeostasis also occurs in sporadic ALS cases. It is well known that these aggregations contain various ubiquitinated components [9]. In addition, ubiquilin2 (UBQLN2), valosin-containing protein (VCP), and optineurin(OPTN), which areassociated with the proteolysis system, have also been identified in as causal genes of ALS [10,11,12]. Thus, protein mislocalization, aggregation formation, and defects in proteolysis could be the fundamental causes of ALS. However, there are many unresolved questions regarding these dysregulations of protein homeostasis (for example, their mechanisms, effects and significance in ALS pathology). Therefore, elucidating such questions would provide the possibility of developing ALS treatments.

    The study of ALS has been steadily increasing, but the exact mechanisms of its pathology and effective treatments remain undiscovered. It isimportant to reveal whether the causal genes of familial ALS share the common motoneuron toxicity with sporadic cases. In this review, we will focus on recent studies of the aberrant distribution of proteins, aggregate formation, and the aberrant proteolysis in familial ALS. In addition, we discuss the possible involvement in sporadic cases.


    2. SOD1


    2.1. SOD1 mutation and ALS

    SOD1 was the first ALS causal gene identified, and approximately 180 ALS-associated mutations have been reported to date [13]. SOD1 contains 153 amino acids and mainly exists in the cytosol, and it is abundant in neuronal cells, including motoneurons. SOD1 forms a homodimer, and each subunit contains one copper ion and one zinc ion. The main function of SOD1 is to eliminate reactive oxygen species, thus it was initially believed that changes in the enzymatic activity of SOD1 by ALS-related mutations caused motoneuron toxicity. However, the enzymatic activity of SOD1 mutants did not correlate with the severity of the ALS phenotype [14,15]. Actually, several mutants of SOD1 maintain their enzymatic activity compared with wild-type SOD1 [16]. Moreover, while SOD1 mutant transgenic mice demonstrate ALS-like phenotypes, SOD1 knockout mice do not [17]. These findings indicate that the motoneuron toxicity of SOD1 mutants was not caused by changes in enzymatic activity but by a gain of toxic functions. As one of the potential mechanisms, it was reported that misfolded SOD1 exerts toxicity through the formation of aggregates and/or the inhibition of cellular functions. Notably, several groups have generated antibodies specific for ALS-associated SOD1 mutants [18,19], and these antibodies recognized structurally abnormal SOD1 not only in SOD1-related but also in sporadic ALS patients [20]. Thus, the abnormal structural changes of SOD1 and subsequent motoneuron toxicity could be a common phenomenon in both familial and sporadic ALS (Figure 1).

    Figure 1.Overview of the potential pathogenic mechanisms of SOD1 toxicities in ALS. The proposed toxicities of SOD1 mutants in SOD1-related ALS. SOD1 mutants form aggregates, which could be prevented by the ER-localizing chaperon PDI in ER. The proper ER functions are inhibited by SOD1 mutants through interactions with Derlin-1 and BiP, resulting in ER stress. SOD1 mutants also abnormally localize in the mitochondrial outer membrane and inhibit the interaction between VDAC and Bcl-2. Chromogranin mediates the extracellular release of SOD1 mutants, triggering microgliosis

    2.2. SOD1 aggregation and post-translation modification

    In histopathological studies, SOD1-positive Lewy body-like inclusions were detected in the ventral horn of the spinal cord in familial ALS patients [15]. Moreover, inclusions containing SOD1 were also observed in the spinal cord of SOD1 mutant transgenic mice in the late stages of the disease [15,21]. It is known that some SOD1-positive inclusions are poly-ubiquitinated and fibrillized [22,23]. When an abnormal protein forms a fiber and eventually an aggregate, it functions as a ‘seed’. The seed gradually gathers proteins that surround it in a process called ‘seeding’, and this process has been reported in SOD1 mutants. Interestingly, a SOD1 seed can induce seeding in cultured cells, and insoluble SOD1 inclusions have been observed [24]. SOD1 mutants associated with ALS have a low stability, and this lower stability causes an abnormal accumulation of SOD1 mutants [25]. Post-translational modifications, metallation and formation of disulfide bonds play a crucial role in maintaining the proper SOD1 conformation. In other words, the proper structure of SOD1 is obtained by the formation of disulfide bonds and metallation of copper and zinc atoms.

    SOD1 contains four cysteine residues (Cys6, 57, 111 and 146), and the intramolecular disulfide bond between Cys57 and Cys146 contributes to the stabilization of SOD1 [26,27]. Indeed, the loss of an intramolecular disulfide bond has been shown to cause SOD1 misfolding [28]. Additionally, oxidative conditions in the ER were suggested to contribute to the formation of disulfide bonds with SOD1. There are over 20 enzymes in the ER that are involved in the oxidization and reduction of proteins. Ero1 is one of the enzymes engaged in the formation of disulfide bonds in the ER, and it strongly interacts with the protein disulfide isomerase (PDI). The Ero1-PDI complex forms a redox network [29] and facilitates the proper structure of proteins through the formation of disulfide bonds. Recently, it was reported that inhibition of PDI catalytic activity promotes SOD1 aggregation [30]. Moreover, a decrease in PDI catalytic activity by S-nitrosylation leads to the formation of SOD1 aggregates in ALS [31,32]. These reports indicate that PDI may help prevent the formation of SOD1 aggregates. This is supported by the finding that PDI co-localizes with SOD1 in familial ALS patients and with TDP-43 in sporadic ALS patients [33]. Thus, it is anticipated that some SOD1 mutants lose their disulfide bonds, which results in abnormal structures that form insoluble and fibrillated aggregates. However, cross-linking of SOD1 through irregular intermolecular disulfide bonds is also a suspected mechanism by which SOD1 forms inclusions [34] (Figure 1).

    Although copper ions are required for substantial SOD1 enzymatic activity, zinc ions increase the stability of SOD1. Notably, mutations in the zinc binding site of SOD1 (H80R, D83G) were reported as causes of ALS. It is also known that the affinity to zinc is reduced in some SOD1 mutants that are prone to forming aggregations [35,36]. Furthermore, a zinc deficiency induces a conformational change of wild-type SOD1 and aberrant SDS-resistant disulfide-linked complex formation [37].

    Considering these previous findings, the abnormal post-translation modifications of SOD1 lead to the formation of aggregates, and the aberrant metabolism of SOD1 can provide the mechanisms by which SOD1 mutants exert toxicity in ALS.


    2.3. Aberrant localization of SOD1 mutants and its toxicities

    SOD1 mutants have been reported to exert the motoneurons toxicities through the mislocalization to extracellular space, mitochondria and ER. The interaction with Chromogranin, a neuronal secretory protein, causes the extracellular secretion of SOD1 mutants. Chromogranin was suggested to act as a chaperone that promotes the secretion of SOD1 mutants through the secretory pathway [38] (Figure 1). The extracellularly secreted SOD1 mutants trigger microgliosis and neurotoxicity.

    Recently macrophage migration inhibitory factor (MIF) was identified as the chaperon that mediates the mislocalization of SOD1 mutants to the outer membrane of mitochondria [39]. SOD1 mutants in mitochondria induce the conformational change of Bcl-2 and inhibit the interaction between Bcl-2 and Voltage-dependent anion channel (VDAC), resulting in the cytotoxicity [40].

    SOD1 mutants also accumulate in the ER and inhibit ER functions [41]. Induction of ER stress, which is an early event in ALS [42], was commonly observed in sporadic ALS patients and SOD1-related ALS animal models [30,41,43]. SOD1 mutants cause ER stress by disrupting protein folding in the ER and endoplasmic reticulum-associated degradation (ERAD), which degrades unfolded proteins in the ER. SOD1 mutants interact with BiP, an ER-resident chaperon, or Derlin-1, a component protein of the ERAD complex [41,44]. Because the induction of ER stress and conformational changes in SOD1 are also observed in sporadic ALS cases [22,45], SOD1-mediated ER stress could also be involved in other types of ALS in addition to SOD1-related ALS.


    2.4. Recent studies of SOD1-mediated motoneurons toxicities using iPSC

    For a long time, the studies of SOD1-related ALS have been heavily dependent on the analyses of overexpressed SOD1 mutants including ALS model animals. In recent years, induced pluripotent stem cells (iPSCs) derived from ALS patients have become a powerful tool. A SOD1-related ALS patient derived iPSCs enables us to analyze the toxicity of SOD1 mutants at the endogenous expression level in human motoneurons. At the beginning, it was reported that iPSCs generated from SOD1-related ALS patient could be differentiated into motoneurons and glial cells [46]. In motoneurons differentiated form SOD1-related ALS patient derived iPSCs, the activation of ER stress, the mitochondrial dysfunction and the accumulation of neurofilament were observed as well as in the SOD1-related ALS patients [47,48]. Furthermore, the recent advantage in genome editing clearly showed this abnormality depends on SOD1 mutation, indicating the usability of iPSC-derived motoneurons as a new ALS model.


    3. TDP-43


    3.1. TDP-43 proteinopathy

    In 2006, TDP-43 was identified in the abnormal protein aggregates that occur in ALS patients [8], and mutations of TARDBP, which encodes TDP-43, have been shown to cause ALS [2]. Notably, the TDP-43-positive aggregation and mutations of TARDBP were found in both familial and sporadic ALS [2]. TDP-43 is also a component of the ubiquitin-positive inclusion found in other neurodegenerative diseases, especially frontotemporal lobar degeneration (FTLD). FTLD presents with progressive neurodegeneration, progressive non-fluent aphasia and sematic dementia. We can classify FTLD pathologically into several subtypes, and one of these subtypes shows motoneuron degeneration similar to that in ALS and is characterized by the ubiquitin- and TDP-43-positive inclusion [49]. The aggregation of TDP-43 is also observed in patients suffering from dementia. Co-localization of tau-protein and α-synuclein were reported in both Alzheimer’s disease and Parkinson’s disease [50]. Although these reports suggest that TDP-43 is widely associated with various neurodegenerative diseases, the detailed molecular mechanisms of TDP-43 pathology remain unclear.


    3.2. TDP-43 function and mutants

    TDP-43 was originally identified as a transcriptional repressor that binds to the TAR DNA of human immunodeficiency virus type 1 [51]; however, subsequent studies revealed that TDP-43 has multiple functions in RNA processing, and it is closely associated with hnRNP (e.g., transcription, splicing, transport, and localization). It was also reported that TDP-43 could stimulate dendritic growth and synapse formation in Drosophila [52], suggesting that TDP-43 is involved at least partially in the maintenance and growth of neurons. TDP-43 contains a nuclear localization signal (NLS), a nuclear export signal (NES), two RRM (RNA-recognition motifs, RRM1 and RRM2) and a glycine-rich domain [51]. Mutagenesis studies showed the presence of functional NLS and NES sequences in TDP-43, and the mutations of NLS and NES induced a restricted distribution of TDP-43 to the cytoplasm and nucleus, respectively [53]. RRM1 is necessary for binding to RNAs and usually recognizes the UG repeat region of RNAs. RRM2 is considered indispensable for interactions with RNAs and is involved in the formation of the TDP-43 dimer; however, its precise role remains unclear [54]. The C-terminal glycine-rich domain is essential for the interaction with some RNA binding proteins and the splicing activity of TDP-43 [55]. Interestingly, over 30 mutations of TARDBP reported in ALS are clustered in the glycine-rich domain [56], indicating the importance of the C-terminal region in ALS pathogenesis. An examination using domain deletion mutants revealed that a part of RRM1 and the C-terminal region were also essential to exerting toxicity in yeast [57].


    3.3. Aberrant post-translational modification of TDP-43

    As mentioned above, TDP-43 was identified as the main component of cytoplasmic inclusions, and various mutations of TDP-43 were found in ALS. Moreover, aberrant post-translational modifications of TDP-43 were found only in inclusions. In a biochemical analysis, the phosphorylation of S409/410 was identified as the major modification of TDP-43 aggregates [58,59]. These findings allowed the distinction between aggregate-forming and soluble TDP-43, but the mechanism by which phosphorylated TDP-43 accumulates in the cytoplasm is still unclear. At the same time, it was also revealed that TDP-43 is cleaved abnormally into a 17- to 25-kDa C-terminal fragment (CTF) (Figure 2). TDP-43 cleavage is mediated by caspase-3, which is activated in apoptotic conditions and in the late stage of the disease [60,61]. Most inclusions, including the pathological 25-kDa CTF, are recognized by the phosphorylated TDP-43-specific antibody [58,62]. However, phosphorylation is not necessary for the cleavage, accumulation and toxicity of TDP-43 [58,62], and only a correlation between the degree of phosphorylation and insolubility has been reported [60]. Although the relationship between the toxicity of the phosphorylated-CTF aggregation and its effect on disease progression is still unclear, the pathological characteristics of TDP-43 proteinopathy (e.g., cytoplasmic accumulation, insolubility, over-phosphorylation, poly-ubiquitination, and abnormal toxicity) are present in cells expressing the 25-kDa CTF [60]. Additionally, in transgenic mice expressing the wild-type TDP-43, production of the 25-kDa CTF increased correspondingly with the disease progression, indicating a crucial role of CTF for motoneuron toxicity [64].

    Figure 2.Overview of TDP-43 and FUS toxicity through aberrant localization and aggregation. (Left) TDP-43 mainly localizes in the nucleus, playing a role in RNA metabolism. TDP-43 shuttles between cytoplasm and nuclear, and mutation or dysregulation of the amount causes the aggregation formation of TDP-43 in cytoplasm. TDP-43-positive aggregations contain an abnormally cleaved and phosphorylated C-terminal fragment (CTF). Seeds and aggregates of TDP-43 have cell-to-cell transmitting ability. (Right) FUS with mutations in the NLS lost the ability to interact with transportin and is prone to localize in the cytoplasm. The excessive stresses facilitate the formation of the cytoplasmic aggregation. The aggregate formation of ALS causative gene products might cause the loss of functions or the gain of toxicity by sequestering the essential components for cell survival

    3.4. Aggregability of TDP-43 and the toxicity of aggregation

    Although wild-type TDP-43 mainly localizes to the nucleus, ALS-related TDP-43 mutants typically translocate to the cytosol and form aggregates in the early stages of ALS [65]. The C-terminal glycine-rich domain of TDP-43, where almost all disease-associated TARDBP gene mutations are clustered, plays an important role in mediating protein-protein interactions. Notably, this domain shows a similarity to yeast prion protein [66], and TDP-43 possesses a prion-like propagation ability. Insoluble TDP-43 isolated from patients with TDP-43 proteinopathy serves as the seed of the aggregation, accumulating the endogenous intact TDP-43 in abnormal aggregations. Furthermore, this seed has cell-to-cell transmitting ability and stability against heating, detergent and proteolysis [67] (Figure 2). ER stress is also suggested to be involved in the formation of TDP-43 aggregates [68]. As one possibility, weak ER stress in the early stage of neurodegenerative disease is reported to induce TDP-43 dysfunction and modulate its distribution. These impairments of TDP-43 then lead to greater ER stress through prion-like propagation and subsequent stabilization of aggregates. Thus, induction of ER stress could facilitate the accumulation of TDP-43 and its toxicity.

    The overexpressed wild-type TDP-43 develops TDP-43 aggregates and cell toxicity, suggesting that dysregulation of the amount of TDP-43 in the cell causes TDP-43 toxicity [64] (Figure 2). It is possible that abnormal accumulation of TDP-43 results from a dysfunction in proteolysis. Indeed, VCP mutation causes the formation of TDP-43 aggregates [10], suggesting that an abnormality in the ubiquitin-proteasome system causes the aggregation. Additionally, it is known that TDP-43, which plays a role in RNA metabolism, autoregulates its expression level [69]. Moreover, it is noteworthy that ALS-related TDP-43 mutants have longer half-lives than the wild-type TDP-43, which correlates with the severity of ALS [70]. Therefore, it is possible that upregulation of TDP-43 through a dysfunction of the degradation system or its induction results in the propagation and subsequent stabilization of the TDP-43 aggregates, which leads to motoneuron death.


    3.5. Studies of animal models and the possible toxicity of TDP-43

    It is still unclear whether TDP-43-mediated neurotoxicity is caused by a loss or gain of function. Therefore, the toxicity of the aberrant localization and aggregation of TDP-43 in motoneurons has been a target of investigation, and several analyses of TDP-43 aggregation models have been attempted. The motoneuron-specific TDP-43 knockout mice exhibited selective and progressive ALS-like motoneuron degeneration, as well as morphological changes in the spinal cord [71]. However, these knockout mice showed different phenotypes than those of ALS, which included the absence of TDP-43 inclusions and survival of upper motoneurons. Because the precise function of TDP-43 in the brain is still unclear, these findings need further study and consideration. In contrast, transgenic mice carrying ALS-associated TDP-43 mutations have a shorter life span than non-transgenic mice and show ALS-like motoneuron degeneration [72]. In addition, transgenic mice overexpressing the wild-type human TDP-43 showed degeneration of cortical and spinal motoneurons, and the level of toxicity in these motoneurons correlated with the expression of TDP-43 [64] (Figure 2). These studies suggest that disturbances in the ability to regulate the amount of TDP-43 causes neurodegeneration, and both the loss of function and gain of toxic function could be mechanisms by which TDP-43 induces cell death.

    The analysis of the possible toxicity of TDP-43 was also performed in smaller animal models such as yeast and Drosophila. In the Drosophila model, the loss of TDP-43 resulted in impairment of locomotive behaviors and life span [73]. Recently, it was also reported that the loss of TDP-43 impaired axonal transport of RNA granules [74] and TDP-43 mutant blocks autophagy-lysosome fusion in Drosophila [75]. In yeast, the modifiers of TDP-43 toxicity were searched through the genome-wide loss of function screenings. The strongest suppression was achieved by the deletion of DBR1, which is a debranching enzyme of lariat introns [76]. This protective effect was confirmed in human neuronal cells or rat primary neuron, and the accumulated lariat introns in DBR1 knocked down cells were suggested to sequester the toxic TDP-43. Another group performed expression screening to identify genes that modulate TDP-43 toxicity also in yeast. They identified PBP-1, an orthologue of human ATXN2, as a TDP-43 toxicity enhancer. This result was confirmed in Drosophila model expressing human wild-type TDP-43, resulting that unregulated Drosophila homologue Atx2 enhanced toxicity of TDP-43 [77]. These results suggested that the fundamental toxicity of TDP-43 would be conserved in small animals such as yeast and Drosophila. Thus, these models in which it is comparatively easy to perform the comprehensive analysis are valuable to reveal the molecular mechanisms and the modifiers of TDP-43 toxicity.

    It is believed that several burdens (e.g., ER stress, genetic impairment of TDP-43, disruption of protein degradation) in cells facilitate the aggregation and stabilization of TDP-43. Because TDP-43 aggregations have been reported to include nonspecific RNAs and RNA binding proteins, the formation of aggregates could exhibit cell toxicity by isolating the essential factors for RNA metabolism. In contrast, a reduction in functional TDP-43 through mutations in TARDBP or other causal genes could suppress the original neuroprotective function of TDP-43. Although the functions of TDP-43 in the nuclear have been taken the notice, recently the importance of cytoplasmic TDP-43 was also highlighted. Cytoplasmic TDP-43 is usually co-localized with stress granules or RNA granules, suggesting that cytoplasmic TDP-43 also has a function in local RNA metabolism [74,78]. Moreover, TDP-43 has been shown to transport the RNA granules along with the axon in the Drosophila model, and the ALS-related mutants lost this ability [74]. These data indicate that the role of cytoplasmic TDP-43 in mRNA metabolisms could also be important in ALS pathogenesis. Further studies are needed to understand the precise mechanisms of TDP-43-mediated motoneuron toxicity.


    4. FUS


    4.1. FUS and neurodegenerative diseases

    Fused in sarcoma (FUS), which is also known as translocated in liposarcoma (TLS), was identified as a fusion oncogene in 1993 [79,80]. In human myxoid liposarcomas, the N-terminus of FUS fuses to a transcriptional factor, leading to an aberrant chromosomal translocation [79,80]. In 2009, FUS was reported as a causal gene of familial ALS, and the relationship between FUS and neurodegenerative diseases was suddenly noticed by researchers. At the same time, the pathological deposits of FUS was reported in FUS-related familial ALS and in FTLD without a FUS mutation [81,82,83].

    FUS belongs to the FET family, which contains FUS, Ewing’s sarcoma breakpoint region 1 (EWS) and TATA-binding protein-associated factor 15 (TAF-15). These three proteins have similar domains and regulate the expression of multiple genes [84]. It was reported that the mutation of TAF15 is associated with ALS, and its mechanism resembles FUS [85]. FET family proteins are involved in not only gene expression but also RNA processing. Although the precise neuronal toxicity is unclear, it is possible that the mutation- or aggregation-mediated dysfunction of RNA maturation mediates neuronal degeneration.


    4.2. Aberrant localization of FUS and aggregation

    FUS contains an N-terminal SYGQ-rich domain, multiple nucleic acid domains and a C-terminal NLS. The NLS region may be the most important region for the physiological function of FUS. Indeed, many FUS mutations that exist in the NLS region have been associated with ALS. transportin, which is a nuclear transport receptor, recognizes the NLS region, and the NLS region is required for nuclear localization of FUS [86,87]. Mutations of FUS (P525L) in the NLS facilitated cytoplasmic redistribution, thus leading to the formation of FUS-positive aggregates [87] (Figure 2). Notably, a mutant’s preference for the cytosol was reported to correlate with the onset of ALS [87]. Moreover, it is well known that the lack of arginine methylation in the RGG3 domain of FUS occurs in FTLD [88]. This defect in arginine methylation causes a tight interaction between FUS and transportin [86,87], and excessive interactions between FUS and transportin impairs the nuclear localization of FUS. Therefore, arginine methylation is required for nuclear translocation of FUS. These lines of evidence suggest that the disturbance in FUS nuclear localization is probably the key step for the formation of cytoplasmic inclusions and motoneuron cell death, which lead to ALS pathogenesis.

    The necessity of a distinct mechanism that regulates the redistribution and aggregation of FUS was suggested. Cytoplasmic FUS is recruited to cytoplasmic deposits, and heat shock or oxidative stress then facilitates the formation of comparatively large inclusions [87] (Figure 2). It is thought that the formation of aggregations containing FUS requires two steps, which are the redistribution of FUS and an excessive amount of cell stress.

    The FUS-positive inclusion was also observed in the brain of FTLD patients without FUS mutations [83], suggesting that FUS mutations are not the sole cause of the formation of FUS inclusions. There are some differences between the FUS-induced pathology in ALS and FTLD. In ALS, the FUS mutations cause impairment in nuclear import, whereas hypomethylation might be the most important step in FTLD. Consistent with these differences, the genetic defect in the FUS gene is always observed in FUS-inclusion-positive ALS, but in FTLD, FUS inclusions are detected even in the absence of a FUS mutation [83,89,90]. Additionally, FUS-specific granules are found in ALS, whereas FUS inclusions in FTLD contain other FET family proteins and transportin, which is required for nuclear import [91,92].

    The toxicity of FUS likely results from the loss of functions in the nucleus and the gain of toxic functions through the aggregates in the cytoplasm. As with TDP-43, analyses involving animal models were performed with FUS to elucidate the source of its toxicity. FUS knockout mice do not show neuronal defects [93,94]. However, motoneuron defects caused by FUS depletion have been reported in zebrafish, which can be rescued by the expression of wild-type FUS [95]. In contrast, human wild-type FUS transgenic mice show a failure to gain weight and paralysis accompanied by impaired motoneuron function [96]. These animal model studies suggest that both the loss of protective functions in the nucleus and the gain of toxic functions through aggregates contribute to the FUS-mediated neurodegenerative pathology.


    5. C9ORF72 toxicity in ALS


    5.1. C9ORF72 mutation in ALS

    The GGGGCC (G4C2) hexanucleotide repeating expansion in the intron region of chromosomal 9 open reading frame 72 (C9ORF72) was reported as a cause of ALS in 2011. Additionally, the abnormal expansion in C9ORF72 was also found in FTLD patients [3,4]. According to the epidemiological study, this abnormal repeating expansion was found in approximately 40% of familial ALS patients [5]. Remarkably, the C9ORF72 mutation was also found in sporadic ALS cases, accounting for approximately 7% of sporadic ALS cases. In normal subjects, the G4C2 hexanucleotide is repeated two to three times, and this sequence is never repeated more than twenty times. However, it is believed that an abnormal expansion is repeated hundreds or even thousands of times in patients with C9ORF72-associated ALS.


    5.2. Motoneuron toxicity resulting from the loss of function mechanism

    Although the precise function of the C9ORF72 encoding protein is still unknown, it was shown that C9ORF72 is a homologue of differentially expressed in normal and neoplasia (DENN), a GDP/GTP exchange factor that activates Rab-GTPase [97], and it is involved in the endosomal trafficking [98]. Thus, mutations of C9ORF72 are considered to disrupt vascular trafficking. C9ORF72 gene is transcribed as three distinct variants and abnormal repeating expansion of variant 2 locates in the promoter region, decreasing its transcription [3]. The decrease of C9ORF72 expression level is reported also in iPSC neurons [99]. To determine whether the loss of function of C9ORF72 causes the motoneurons toxicities, several animal models were generated. While some of these model animals (C. elegans and zebrafish) showed neuronal deficit and dysfunctional behavior [100,101], neural-specific ablation of C9ORF72 in mice does not result in the ALS-like phenotype [102]. Thus, it is still unclear whether the loss of function of C9ORF72 causes motoneuron degeneration and further studies are required.


    5.3. Motoneuron toxicity resulting from the gain of function mechanism

    The abnormal RNA is considered to gain toxicity. The toxicity of abnormal RNA repeat expansions can be observed in several diseases. For example, in muscular dystrophy, which shows muscular necrosis and multiple organ failure, abnormal RNA repeats sequester essential proteins for RNA processing in aggregations such as RNA foci, leading to disruption of RNA metabolism and cell death [103]. In the case of C9ORF72-mediated ALS, RNA foci are observed in neurons derived from iPSC as well as in spinal and frontal cortex neurons of ALS patients [3,99]. It has been proposed that C9ORF72 RNA containing a G4C2 repeat expansion also sequesters essential proteins and disturbs the RNA metabolism. The interactions with ribonucleoproteins like Pur-α and ADARB2 are considered to cause nucleolar stress [99,104,105].

    A toxicity of abnormally expanded dipeptide repeats was also reported. Translation independent of the start codon (ATG) is induced in the presence of abnormal repeat expansions (repeat-associated non-ATG (RAN) translation) [106]. Theoretically, there are five expression patterns of the dipeptide repeat (DPR) protein generated through the RAN translation. Among them, the strong Gly-Ala DPR-positive inclusions, and to a lesser extent, the Gly-Pro and Gly-Arg DPRs-positive inclusions, were found in the brains of ALS and FTLD patients [107]. Notably, aggregations of DPRs were found together with TDP-43 negative/p62 positive inclusions, thus suggesting TDP-43-independent toxicity [3,107,108,109]. Several groups have demonstrated the toxicity of C9ORF72 DPRs in various models, indicating the importance of DPRs in neurodegeneration [107,109]. The toxicity of each DPRs are considered to differ from each other, and the analysis of the Gly-Arg DPR toxicity has been promoted. The expression of Gly-Ala DPR has been reported to evoke ER stress through activating PKR-like endoplasmic reticulum kinase (PERK) pathway [110]. Since other ER stress sensors were not activated by Gly-Ala DPR, the mere inhibition of ubiquitin proteasome system, one of proposed mechanism, was not likely and further analyses are needed to identify the specific target of Gly-Ala DPR.

    To determine which of the abnormal RNA or DPR is the main cause of motoneuronal toxicity of C9ORF72 mutation, two different fly models were generated. The one line has long repeat RNA with stop codons, which generates only RNA repeat expansion and not DPRs. The other line generates, besides RNA repeats, DPRs from RAN translation [111]. The line expressing both RNA repeats and DPRs showed toxicity, but the line expressing only RNA repeats had little effect. The other group generated fly model with a C9ORF72 minigene containing G4C2 repeats [112]. This fly model showed RNA foci in nuclear but not significant toxicities. These reports utilizing small animals indicated that DPR is the major toxic species compared to abnormal RNA. Recently, several groups have also generated C9ORF72-ALS model mice with distinct techniques. One group generated mice carrying Bacterial artificial chromosome (BAC) harboring full-length human C9ORF72 containing G4C2 repeat expansion. Despite RNA foci and DPR from RAN translation were observed in these mice, aberrant behavior and neurodegeneration were not [113]. The other group used adeno-associated virus (AAV) containing 66 G4C2 repeats and administered these viruses to postnatal day 0 mice. Six-month-old mice exhibit besides accumulation of RNA foci and DPRs, neuronal loss and gliosis [114]. Though several C9ORF72 mutation models are evaluated, further studies are required to conclude the mechanism by which C9ORF72 mutation causes toxicity. Notably, two recent studies suggested the involvement of a defect in nucleocytoplasmic transport in C9ORF72 mutation-induced toxicity [115,116]. In Drosophila screening, RanGAP1 was identified as a suppressor of C9ORF72 mutation-induced neuronal toxicity. They demonstrated the physical interaction of RanGAP1 with an RNA repeat expansion produced by G4C2 repeat. This interaction might cause inhibition of RanGAP1 and aberrant nucleocytoplasmic distribution [115]. Another report demonstrated that a wide variety of nuclear pore proteins could modulate C9ORF72 mutation-induced toxicity, and the dysfunction of nucleocytoplasmic transport facilitated the accumulation of RNA in the nucleus. Moreover, it was reported that C9ORF72 DPR toxicity is also modified by nucleocytoplasmic transport [117] (Figure 3).

    Figure 3.Overview of C9ORF72 toxicity through repeat expansion of RNA and DPRs. The intronic hexanucleotide repeat expansion in C9ORF72 causes the formation of RNA foci in the nuclear. RNA foci might sequester RNA binding proteins that are essential for cell survival. Dipeptide repeat proteins (DPRs) are generated from aberrant repeat expansion through RAN translation, evoking ER stress. DPR-positive aggregation is colocalized with p62 in the cytosol. The interaction of abnormal RNA repeat expansion with ribonucleoproteins is considered to cause nucleolar stress. A defect in nucleocytoplasmic transport has also been reported as the toxicity of the C9ORF72 mutation

    It is still unclear how C9ORF72 mutation causes toxicity through repeat expansion RNA or the production of DPRs, or both, the dysfunction of nucleocytoplasmic transport and subsequent aberrant distribution of proteins or RNAs could be the cause of motoneuron toxicity.


    6. Other causal genes of ALS


    6.1. OPTN

    In 2010, OPTN, which is a known risk gene for primary open-angle glaucoma, was also identified as a causal gene for ALS [12,118]. OPTN contains 577 amino acids and has a coiled-coil domain, zinc-finger domain and ubiquitin-binding domain. OPTN has multiple functions, and its main function is negative regulation of NF-κB activation. It competes with NF-κB essential modulator (NEMO) for binding to ubiquitinated receptor interacting protein (RIP) to prevent NF-κB activation [119]. The ALS-associated OPTN E487G mutant, which has a mutation in the ubiquitin-binding domain, has a loss of function for binding to ubiquitin that prevents inhibition of NF-κB activation [120]. It is likely that the functional disruption of OPTN induces an aberrant activation of NF-κB, which leads to neurodegeneration. OPTN E487G mutant shows aberrant cell localization and formation of cytosolic inclusions. In addition, OPTN has the functions of maintaining the Golgi body and transport from the Golgi body [121]. Moreover, OPTN also possesses the role of an autophagy receptor. OPTN localizes with the LC3 vesicle, and its ability to bind with ubiquitin is necessary for adapting to LC3. The OPTN E478G mutant loses the ability to bind to ubiquitin as well as to co-localize with LC3 [122]. OPTN is also detected in ubiquitin- and TDP-43-positive inclusions in sporadic ALS, in SOD1-positive inclusions in familial SOD1-related ALS, and in FUS-positive inclusions in FUS-related ALS [119,123,124]. These results suggest that OPTN-positive inclusion is a common phenomenon in wide range of ALS types. Although the exact toxicity is unknown, the sequestration of OPTN to aggregations and its loss of function could be a common cause of ALS.


    6.2. UBQLN2 and VCP

    UBQLN2, which is associated with the ubiquitin-proteasome system, was also identified as a causal gene of ALS. In X-linked ALS-FTD, UBQLN2 had several mutations in the PXX repeat domain [11]. Recent studies also reported mutations in the vicinity of the PXX domain as a cause of ALS [125]. Although the precise function of the PXX domain is still unclear, UBQLN2 may play a role in ubiquitin-dependent proteolysis and the mutation might result in the dysfunction of protein degradation [11,126]. UBQLN2-positive inclusion was observed in ALS-FTD patients with or without UBQLN2 mutations [11]. Skein-like inclusions are positive for UBQLN2 in the spinal cords of ALS patients with SOD1, TARDBP and FUS mutations [11,125].

    The gene encoding VCP was also reported as a causal gene of ALS, and VCP mutation accounts for 1-2% of familial ALS cases [10]. It was shown that VCP interacted with TDP-43 and that disease-causing VCP mutants led to the redistribution of TDP-43 to the cytoplasm [127]. VCP forms a complex with its co-factors, Npl4 and Ufd1, and interacts with ubiquitinated proteins or ERAD complex, such as Hrd1 and Derlin-1 [128,129,130,131]. In ERAD, unfolded proteins in the ER are transported from the ER to the cytosol and subsequently degraded by the ubiquitin-proteasome system in the cytosol. It is suggested that VCP contributes to the retrograde-translocation of unfolded proteins through its ATPase activity [129,130]. Notably, UBQLN2 is also estimated to be involved in ERAD. UBQLN2 has a ubiquitin-like domain and a ubiquitin-associated domain and is considered to facilitate the transport of ubiquitinated substrates to proteasomes [126]. As mentioned before, SOD1 mutants also inhibit ERAD. Thus, it is possible that the dysfunction of ERAD, which leads to the defect in proteolysis, commonly plays an important role in ALS pathogenesis.


    7. Conclusions

    The pathological mechanism of ALS is still unclear, and there is no fundamental or effective therapy. Many causal genes of ALS have recently been identified. Numerous studies analyzing these genes have demonstrated multiple forms of motoneuron toxicity in each mutation-induced ALS. However, there is a need to reveal common mechanisms of familial ALS pathogenesis to elucidate the pathogenesis of sporadic ALS, which accounts for 90% of all cases. In order to approach the common toxicity in ALS motoneurons, some important questions below should be solved.


    7.1. Whether the ALS model animals really represent the ALS pathology

    Along with the discovery of ALS causative genes, there has been much effort to generate ALS model animals. Through the generation of knockout or transgenic animals of ALS-related genes, some animal models that show the motoneuronal degeneration and ALS-like symptoms have been established. While some of them possess high similarity to the ALS, the complete knockout or the excessive expression of ALS-relate genes could cause artificial toxicities; but we should carefully interpret the results of these animal models. Especially, the pathological phenomenon that we focus on this review would be affected through the difference of expression levels. Since animal models give us significant information and are powerful tools to analyze in vivo effects, the confirmation in higher animals is desired. The generation of the knock-in animals of ALS-related mutation might also be useful. In addition, motoneurons differentiated from iPSC derived from ALS patients have been reported to reproduce the motoneurons toxicities [46,132,133]. In this model, the toxicity of each mutation could be assayed in the physiological conditions. Although there are several issues to be addressed (stability, clonal effect, lack of non-cell autonomous effects), iPSCs may also become prominent ALS model.


    7.2. The problem of species difference

    One of the big problems to develop the ALS treatment is that drugs effective in ALS model animals do not exert enough effect on ALS patients. The model animal could be unsuitable to evaluate the human ALS, which appears to be caused in part by the species difference. Although the animal models share the same toxicities with human ALS, most of the drugs target the motoneurons toxicity but not the fundamental mechanism of pathogenesis, which should be targeted to solve the problems of the species difference. Thus, the elucidation of the precise pathogenesis of ALS and discovery of molecular mechanism-based ALS treatment agent are required. Also, the iPSC-ALS model would be useful, in which we can evaluate the treatment efficiency in human.


    7.3. Whether ALS cases share the common pathogenesis

    An important question to deal with ALS (especially to approach sporadic ALS treatment) is whether ALS with different gene mutations is caused by the common mechanism. The symptoms observed in patients and the properties of the autopsy sometimes differ from the others. For example, in SOD1-related ALS the TDP-43 positive inclusion that is frequently observed in other type of ALS patients has not been found [134]. It would be possible that each genetic mutations of ALS exert its toxicity through different mechanisms. On the other hand, SOD1 has also been reported to be involved in the non-cell autonomous motoneurons toxicity in SOD1 mutation-negative sporadic ALS, suggesting SOD1 as the common mediator of ALS toxicities [38,135]. Although the number of cases analyzed for the involvement of SOD1 in sporadic cases is still too small and SOD1 does not necessarily participate in all sporadic cases [134], the further large-scale analyses are desired. It is worth noting that the aberrant distribution of proteins/RNAs and the formation of aggregations were detected in a wide range of ALS types, even in the absence of their mutations. Thus, an analysis of the mislocalization, aggregation formation and defects in proteolysis could provide the insight into the common pathological events in ALS. In addition, the development of iPSC technology would enable us to investigate the molecular mechanisms of sporadic ALS cases. These attempts would reveal the common pathogenesis in wider variety of ALS cases and shed light on the fundamental ALS treatment.


    Acknowledgements

    We would like to thank the members in the laboratory of Cell Signaling who gave us appropriate directions and valuable advice.


    Conflict of interest

    The authors declare that they have no conflict of interest.


    [1] Rosen DR, Siddique T, Patterson D, et al. (1993) Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 362: 59-62. doi: 10.1038/362059a0
    [2] Sreedharan J, Blair IP, Tripathi VB, et al. (2008) TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science 249: 1668-1672.
    [3] DeJesus-Hernandez M, Mackenzie IR, Boeve BF, et al. (2011) Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 72: 245-256. doi: 10.1016/j.neuron.2011.09.011
    [4] Renton AE, Majounie E, Waite A, et al. (2011) A Hexanucleotide repeat expansion in C9ORF72 is the aause of chromosome 9p21-linked ALS-FTD. Neuron 72: 257-268. doi: 10.1016/j.neuron.2011.09.010
    [5] Majounie E, Renton AE, Mok K, et al. (2012) Frequency of the C9orf72 hexanucleotide repeat expansion in patients with amyotrophic lateral sclerosis and frontotemporal dementia: a cross-sectional study. Lancet Neurol 11: 323-330. doi: 10.1016/S1474-4422(12)70043-1
    [6] Keller BA, Volkening K, Droppelmann CA, et al. (2012) Co-aggregation of RNA binding proteins in ALS spinal motor neurons: evidence of a common pathogenic mechanism. Acta Neuropathol 124: 733-747. doi: 10.1007/s00401-012-1035-z
    [7] Neumann M, Sampathu DM, Kwong LK, et al. (2006) Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 130: 130-133.
    [8] Arai T, Hasegawa M, Akiyama H, et al. (2006) TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem Biophys Res Commun 351: 602-611. doi: 10.1016/j.bbrc.2006.10.093
    [9] Leigh PN, Anderton BH, Dodson A, et al. (1988) Ubiquitin deposits in anterior horn cells in motor neuron disease. Neurosci Lett 93: 197-203. doi: 10.1016/0304-3940(88)90081-X
    [10] Johnson JO, Mandrioli J, Benatar M, et al. (2010) Exome sequencing reveals VCP mutations as a cause of familial ALS. Neuron 68: 857-864. doi: 10.1016/j.neuron.2010.11.036
    [11] Deng H-X, Chen W, Hong S-T, et al. (2011) Mutations in UBQLN2 cause dominant X-linked juvenile and adult-onset ALS and ALS/dementia. Nature 477: 211-215. doi: 10.1038/nature10353
    [12] Maruyama H, Morino H, Ito H, et al. (2010) Mutations of optineurin in amyotrophic lateral sclerosis. Nature 465: 223-226. doi: 10.1038/nature08971
    [13] ALSoD (Amyotrophic Lateral Sclerosis Online Genetics Database). Available from: http://alsod.iop.kcl.ac.uk/
    [14] Cleveland DW, Laing N, Hurse PV, et al. (1995) Toxic mutants in Charcot’s sclerosis. Nature 378: 342-343. doi: 10.1038/378342a0
    [15] Bruijn LI, Houseweart MK, Kato S, et al. (1998) Aggregation and motor neuron toxicity of an ALS-linked SOD1 mutant independent from wild-type SOD1. Science 281: 1851-1854.
    [16] Hayward LJ, Rodriguez JA, Kim JW, et al. (2002) Decreased metallation and activity in subsets of mutant superoxide dismutases associated with familial amyotrophic lateral sclerosis. J Biol Chem 277: 15923-15931. doi: 10.1074/jbc.M112087200
    [17] Reaume AG, Elliott JL, Hoffman EK, et al. (1996) Motor neurons in Cu/Zn superoxide dismutase-deficient mice develop normally but exhibit enhanced cell death after axonal injury. Nat Genet 13: 43-47. doi: 10.1038/ng0596-43
    [18] Fujisawa T, Homma K, Yamaguchi N, et al. (2012) A novel monoclonal antibody reveals a conformational alteration shared by amyotrophic lateral sclerosis-linked SOD1 mutants. Ann Neurol 72: 739-49. doi: 10.1002/ana.23668
    [19] Urushitani M, Ezzi SA, Julien JP (2007) Therapeutic effects of immunization with mutant superoxide dismutase in mice models of amyotrophic lateral sclerosis. Proc Natl Acad Sci U S A 104: 2495-2500. doi: 10.1073/pnas.0606201104
    [20] Bosco DA, Morfini G, Karabacak NM, et al. (2010) Wild-type and mutant SOD1 share an aberrant conformation and a common pathogenic pathway in ALS. Nat Neurosci 13: 1396-1403. doi: 10.1038/nn.2660
    [21] Watanabe M, Dykes-Hoberg M, Culotta VC, et al. (2001) Histological evidence of protein aggregation in mutant SOD1 transgenic mice and in amyotrophic lateral sclerosis neural tissues. Neurobiol Dis 8: 933-941. doi: 10.1006/nbdi.2001.0443
    [22] Basso M, Massignan T, Samengo G, et al. (2006) Insoluble mutant SOD1 is partly oligoubiquitinated in amyotrophic lateral sclerosis mice. J Biol Chem 281: 33325-33335. doi: 10.1074/jbc.M603489200
    [23] Wang J, Xu G, Gonzales V, et al. (2002) Fibrillar inclusions and motor neuron degeneration in transgenic mice expressing superoxide dismutase 1 with a disrupted copper-binding site. Neurobiol Dis 10: 128-138. doi: 10.1006/nbdi.2002.0498
    [24] Furukawa Y, Fu R, Deng HX, et al. (2006) Disulfide cross-linked protein represents a significant fraction of ALS-associated Cu, Zn-superoxide dismutase aggregates in spinal cords of model mice. Proc Natl Acad Sci U S A 103: 7148-7153. doi: 10.1073/pnas.0602048103
    [25] Rodriguez JA, Valentine JS, Eggers DK, et al. (2002) Familial amyotrophic lateral sclerosis-associated mutations decrease the thermal stability of distinctly metallated species of human Copper/Zinc superoxide dismutase. J Biol Chem 277: 15932-15937. doi: 10.1074/jbc.M112088200
    [26] Sea K, Sohn SH, Durazo A, et al. (2015) Insights into the role of the unusual disulfide bond in Copper-Zinc superoxide dismutase. J Biol Chem 290: 2405-2418. doi: 10.1074/jbc.M114.588798
    [27] Doucette PA, Whitson LJ, Cao X, et al. (2004) Dissociation of human copper-zinc superoxide dismutase dimers using chaotrope and reductant: Insights into the molecular basis for dimer stability. J Biol Chem 279: 54558-54566. doi: 10.1074/jbc.M409744200
    [28] Hough MA, Grossmann JG, Antonyuk SV, et al. (2004) Dimer destabilization in superoxide dismutase may result in disease-causing properties: structures of motor neuron disease mutants. Proc Natl Acad Sci U S A 101: 5976-5981. doi: 10.1073/pnas.0305143101
    [29] Araki K, Iemura S, Kamiya Y, et al. (2013) Ero1-α and PDIs constitute a hierarchical electron transfer network of endoplasmic reticulum oxidoreductases. J Cell Biol 202: 861-874. doi: 10.1083/jcb.201303027
    [30] Atkin JD, Farg MA, Turner BJ, et al. (2006) Induction of the unfolded protein response in familial amyotrophic lateral sclerosis and association of protein-disulfide isomerase with superoxide dismutase 1. J Biol Chem 281: 30152-30165. doi: 10.1074/jbc.M603393200
    [31] Chen X, Zhang X, Li C, et al. (2013) S-nitrosylated protein disulfide isomerase contributes to mutant SOD1 aggregates in amyotrophic lateral sclerosis. J Neurochem 124: 45-58. doi: 10.1111/jnc.12046
    [32] Jeon GS, Nakamura T, Lee JS, et al. (2014) Potential effect of S-nitrosylated protein disulfide isomerase on mutant SOD1 aggregation and neuronal cell death in amyotrophic lateral sclerosis. Mol Neurobiol 49: 796-807. doi: 10.1007/s12035-013-8562-z
    [33] Honjo Y, Kaneko S, Ito H, et al. (2011) Protein disulfide isomerase-immunopositive inclusions in patients with amyotrophic lateral sclerosis. Amyotroph Lateral Scler 2968: 1-7.
    [34] Toichi K, Yamanaka K, Furukawa Y (2013) Disulfide scrambling describes the oligomer formation of superoxide dismutase (SOD1) proteins in the familial form of amyotrophic lateral sclerosis. J Biol Chem 288: 4970-4980. doi: 10.1074/jbc.M112.414235
    [35] Crow JP, Sampson JB, Zhuang Y, et al. (1997) Decreased zinc affinity of amyotrophic lateral sclerosis-associated superoxide dismutase mutants leads to enhanced catalysis of tyrosine nitration by peroxynitrite. J Neurochem 69: 1936-1944.
    [36] Lyons TJ, Liu H, Goto JJ, et al. (1996) Mutation in copper-zinc superoxide dismutase that cause amyotrophic lateral sclerosis alter the zinc binding site and the redox behavior of the protein. Proc Natl Acad Sci U S A 93: 12240-12244. doi: 10.1073/pnas.93.22.12240
    [37] Homma K, Fujisawa T, Tsuburaya N, et al. (2013) SOD1 as a molecular switch for initiating the homeostatic ER stress response under zinc deficiency. Mol Cell 52: 75-86. doi: 10.1016/j.molcel.2013.08.038
    [38] Urushitani M, Sik A, Sakurai T, et al. (2006) Chromogranin-mediated secretion of mutant superoxide dismutase proteins linked to amyotrophic lateral sclerosis. Nat Neurosci 9: 108-118. doi: 10.1038/nn1603
    [39] Israelson A, Ditsworth D, Sun S, et al. (2013) Macrophage migration inhibitory factor as a chaperone inhibiting accumulation of misfolded SOD1. Neuron 86: 218–232. doi: 10.1016/j.neuron.2015.02.034
    [40] Tan W, Naniche N, Bogush A, et al. (2013) Small peptides against the mutant SOD1/Bcl-2 toxic mitochondrial complex restore mitochondrial function and cell viability in mutant SOD1-mediated ALS. J Neurosci 33: 11588-11598. doi: 10.1523/JNEUROSCI.5385-12.2013
    [41] Kikuchi H, Almer G, Yamashita S, et al. (2006) Spinal cord endoplasmic reticulum stress associated with a microsomal accumulation of mutant superoxide dismutase-1 in an ALS model. Proc Natl Acad Sci U S A 103: 6025-6030. doi: 10.1073/pnas.0509227103
    [42] Sun S, Sun Y, Ling SC, et al. (2015) Translational profiling identifies a cascade of damage initiated in motor neurons and spreading to glia in mutant SOD1-mediated ALS. Proc Natl Acad Sci U S A 112: E6993-E7002. doi: 10.1073/pnas.1520639112
    [43] Ito Y, Yamada M, Tanaka H, et al. (2009) Involvement of CHOP, an ER-stress apoptotic mediator, in both human sporadic ALS and ALS model mice. Neurobiol Dis 36: 470-476. doi: 10.1016/j.nbd.2009.08.013
    [44] Nishitoh H, Kadowaki H, Nagai A, et al. (2008) ALS-linked mutant SOD1 induces ER stress- and ASK1-dependent motor neuron death by targeting Derlin-1. Genes Dev 22: 1451-1464. doi: 10.1101/gad.1640108
    [45] Pokrishevsky E, Grad LI, Yousefi M, et al. (2012) Aberrant localization of FUS and TDP43 is associated with misfolding of SOD1 in amyotrophic lateral sclerosis. PLoS One 7: 1-9.
    [46] Dimos JT, Rodolfa KT, Niakan KK, et al. (2008) Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science 321: 1218-1221. doi: 10.1126/science.1158799
    [47] Chen H, Qian K, Du Z, et al. (2014) Modeling ALS with iPSCs reveals that mutant SOD1 misregulates neurofilament balance in motor neurons. Cell Stem Cell 14: 796-809. doi: 10.1016/j.stem.2014.02.004
    [48] Kiskinis E, Sandoe J, Williams LA, et al. (2014) Pathways disrupted in human ALS motor neurons identified through genetic correction of mutant SOD1. Cell Stem Cell 14: 781-795. doi: 10.1016/j.stem.2014.03.004
    [49] Neary D, Snowden JS, Mann DM (2000) Classification and description of frontotemporal dementias. Ann N Y Acad Sci 920: 46-51.
    [50] Higashi S, Iseki E, Yamamoto R, et al. (2007) Concurrence of TDP-43, tau and α-synuclein pathology in brains of Alzheimer’s disease and dementia with Lewy bodies. Brain Res 1184: 284-294. doi: 10.1016/j.brainres.2007.09.048
    [51] Ou SH, Wu F, Harrich D, et al. (1995) Cloning and characterization of a novel cellular protein, TDP-43, that binds to human immunodeficiency virus type 1 TAR DNA sequence motifs. J Virol 69: 3584-3596.
    [52] Lu Y, Ferris J, Gao FB (2009) Frontotemporal dementia and amyotrophic lateral sclerosis-associated disease protein TDP-43 promotes dendritic branching. Mol Brain 2: 30. doi: 10.1186/1756-6606-2-30
    [53] Winton MJ, Igaz LM, Wong MM, et al. (2008) Disturbance of nuclear and cytoplasmic TAR DNA-binding protein (TDP-43) induces disease-like redistribution, sequestration, and aggregate formation. J Biol Chem 283: 13302-13309. doi: 10.1074/jbc.M800342200
    [54] Kuo PH, Doudeva LG, Wang YT, et al. (2009) Structural insights into TDP-43 in nucleic-acid binding and domain interactions. Nucleic Acids Res 37: 1799-1808. doi: 10.1093/nar/gkp013
    [55] Buratti E, Brindisi A, Giombi M, et al. (2005) TDP-43 binds heterogeneous nuclear ribonucleoprotein A/B through its C-terminal tail: an important region for the inhibition of cystic fibrosis transmembrane conductance regulator exon9 splicing. J Biol Chem 280: 37572-37584. doi: 10.1074/jbc.M505557200
    [56] Kabashi E, Valdmanis PN, Dion P, et al. (2008) TARDBP mutations in individuals with sporadic and familial amyotrophic lateral sclerosis. Nat Genet 40: 572-574. doi: 10.1038/ng.132
    [57] Johnson BS, McCaffery JM, Lindquist S, et al. (2008) A yeast TDP-43 proteinopathy model: Exploring the molecular determinants of TDP-43 aggregation and cellular toxicity. Proc Natl Acad Sci U S A 105: 6439-6444. doi: 10.1073/pnas.0802082105
    [58] Hasegawa M, Arai T, Nonaka T, et al. (2008) Phosphorylated TDP-43 in frontotemporal lobar degeneration and ALS. Ann Neurol 64: 60-70. doi: 10.1002/ana.21425
    [59] Kadokura A, Yamazaki T, Kakuda S, et al. (2009) Phosphorylation-dependent TDP-43 antibody detects intraneuronal dot-like structures showing morphological characters of granulovacuolar degeneration. Neurosci Lett 463: 87-92. doi: 10.1016/j.neulet.2009.06.024
    [60] Igaz LM, Kwong LK, Chen-Plotkin A, et al. (2009) Expression of TDP-43 C-terminal fragments in vitro recapitulates pathological features of TDP-43 proteinopathies. J Biol Chem 284: 8516-8524. doi: 10.1074/jbc.M809462200
    [61] Zhang YJ, Xu YF, Cook C, et al. (2009) Aberrant cleavage of TDP-43 enhances aggregation and cellular toxicity. Proc Natl Acad Sci U S A 106: 7607-7612. doi: 10.1073/pnas.0900688106
    [62] Arai T, Hasegawa M, Akiyama H, et al. (2006) TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem Biophys Res Commun 351: 602-611. doi: 10.1016/j.bbrc.2006.10.093
    [63] Dormann D, Capell A, Carlson AM, et al. (2009) Proteolytic processing of TAR DNA binding protein-43 by caspases produces C-terminal fragments with disease defining properties independent of progranulin. J Neurochem 110: 1082-1094. doi: 10.1111/j.1471-4159.2009.06211.x
    [64] Wils H, Kleinberger G, Janssens J, et al. (2010) TDP-43 transgenic mice develop spastic paralysis and neuronal inclusions characteristic of ALS and frontotemporal lobar degeneration. Proc Natl Acad Sci U S A 107: 3858-3863. doi: 10.1073/pnas.0912417107
    [65] Giordana MT, Piccinini M, Grifoni S, et al. (2010) TDP-43 redistribution is an early event in sporadic amyotrophic lateral sclerosis. Brain Pathol 20: 351-360. doi: 10.1111/j.1750-3639.2009.00284.x
    [66] Udan M, Baloh RH (2011) Implications of the prion-related Q/N domains in TDP-43 and FUS. Prion 5: 1-5.
    [67] Nonaka T, Masuda-Suzukake M, Arai T, et al. (2013) Prion-like properties of pathological TDP-43 aggregates from diseased brains. Cell Rep 4: 124-134. doi: 10.1016/j.celrep.2013.06.007
    [68] Walker AK, Soo KY, Sundaramoorthy V, et al. (2013) ALS-associated TDP-43 induces endoplasmic reticulum stress, which drives cytoplasmic TDP-43 accumulation and stress granule formation. PLoS One 8: e81170. doi: 10.1371/journal.pone.0081170
    [69] Polymenidou M, Lagier-Tourenne C, Hutt KR, et al. (2011) Long pre-mRNA depletion and RNA missplicing contribute to neuronal vulnerability from loss of TDP-43. Nat Neurosci 14: 459-468. doi: 10.1038/nn.2779
    [70] Watanabe S, Kaneko K, Yamanaka K (2013) Accelerated disease onset with stabilized familial amyotrophic lateral ssclerosis (ALS)-linked mutant TDP-43 proteins. J Biol Chem 288: 3641-3654. doi: 10.1074/jbc.M112.433615
    [71] Iguchi Y, Katsuno M, Niwa J, et al. (2013) Loss of TDP-43 causes age-dependent progressive motor neuron degeneration. Brain 136: 1371-1382. doi: 10.1093/brain/awt029
    [72] Wegorzewska I, Bell S, Cairns NJ, et al. (2009) TDP-43 mutant transgenic mice develop features of ALS and frontotemporal lobar degeneration. Proc Natl Acad Sci U S A 106: 18809-18814. doi: 10.1073/pnas.0908767106
    [73] Feiguin F, Godena VK, Romano G, et al. (2009) Depletion of TDP-43 affects Drosophila motoneurons terminal synapsis and locomotive behavior. FEBS Lett 583: 1586-1592. doi: 10.1016/j.febslet.2009.04.019
    [74] Alami NH, Smith RB, Carrasco MA, et al. (2013) Axonal transport of TDP-43 mRNA granules is impaired by ALS-causing mutations. Neuron 81: 536-543.
    [75] Xia Q, Wang H, Hao Z, et al. (2015) TDP-43 loss of function increases TFEB activity and blocks autophagosome-lysosome fusion. EMBO J 35: 1-22.
    [76] Armakola M, Higgins MJ, Figley MD, et al. (2012) Inhibition of RNA lariat debranching enzyme suppresses TDP-43 toxicity in ALS disease models. Nat Genet 44: 1302-1309. doi: 10.1038/ng.2434
    [77] Elden AC, Kim HJ, Hart MP, et al. (2010) Ataxin-2 intermediate-length polyglutamine expansions are associated with increased risk for ALS. Nature 466: 1069-1075. doi: 10.1038/nature09320
    [78] Liu-Yesucevitz L, Lin AY, Ebata A, et al. (2014) ALS-linked mutations enlarge TDP-43-enriched neuronal RNA granules in the dendritic arbor. J Neurosci 34: 4167-4174. doi: 10.1523/JNEUROSCI.2350-13.2014
    [79] Crozat A, Aman P, Mandahl N, et al. (1993) Fusion of CHOP to a novel RNA-binding protein in human myxoid liposarcoma. Nature 363: 640-644. doi: 10.1038/363640a0
    [80] Rabbits TH, Forster A, Larson R, et al. (1993) Fusion of the dominant negative transcription regulator CHOP with a novel gene FUS by translocation t (12; 16) in malignant liposarcoma. Nat Genet 4: 175-180. doi: 10.1038/ng0693-175
    [81] Vance C, Rogelj B, Hortobágyi T, et al. (2009) Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis Type 6. Science 323: 1208-1211. doi: 10.1126/science.1165942
    [82] Kwiatkowski TJ, Bosco DA, Leclerc AL, et al. (2009) Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science 323: 1205-1208. doi: 10.1126/science.1166066
    [83] Neumann M, Rademakers R, Roeber S, et al. (2009) A new subtype of frontotemporal lobar degeneration with FUS pathology. Brain 132: 2922-2931. doi: 10.1093/brain/awp214
    [84] Kovar H (2011) The two faces of the FUS/EWS/TAF15 protein family. Sarcoma 2011: 1-13.
    [85] Couthouis J, Hart MP, Erion R, et al. (2012) Evaluating the role of the FUS/TLS-related gene EWSR1 in amyotrophic lateral sclerosis. Hum Mol Genet 21: 2899-2911. doi: 10.1093/hmg/dds116
    [86] Lee BJ, Cansizoglu AE, Süel KE, et al. (2006) Rules for Nuclear Localization Sequence Recognition by Karyopherinβ2. Cell 126: 543-558. doi: 10.1016/j.cell.2006.05.049
    [87] Dormann D, Rodde R, Edbauer D, et al. (2010) ALS-associated fused in sarcoma (FUS) mutations disrupt Transportin-mediated nuclear import. EMBO J 29: 2841-2857. doi: 10.1038/emboj.2010.143
    [88] Dormann D, Madl T, Valori CF, et al. (2012) Arginine methylation next to the PY-NLS modulates Transportin binding and nuclear import of FUS. EMBO J 31: 4258-4275. doi: 10.1038/emboj.2012.261
    [89] Snowden JS, Hu Q, Rollinson S, et al. (2011) The most common type of FTLD-FUS (aFTLD-U) is associated with a distinct clinical form of frontotemporal dementia but is not related to mutations in the FUS gene. Acta Neuropathol 122: 99-110. doi: 10.1007/s00401-011-0816-0
    [90] Urwin H, Josephs KA, Rohrer JD, et al. (2010) FUS pathology defines the majority of tau-and TDP-43-negative frontotemporal lobar degeneration. Acta Neuropathol 120: 33-41. doi: 10.1007/s00401-010-0698-6
    [91] Brelstaff J, Lashley T, Holton JL, et al. (2011) Transportin1: a marker of FTLD-FUS. Acta Neuropathol 122: 591-600. doi: 10.1007/s00401-011-0863-6
    [92] Davidson YS, Robinson AC, Hu Q, et al. (2013) Nuclear carrier and RNA-binding proteins in frontotemporal lobar degeneration associated with fused in sarcoma (FUS) pathological changes. Neuropathol Appl Neurobiol 39: 157-165. doi: 10.1111/j.1365-2990.2012.01274.x
    [93] Kuroda M, Sok J, Webb L, et al. (2000) Male sterility and enhanced radiation sensitivity in TLS(-/-) mice. EMBO J 19: 453-462. doi: 10.1093/emboj/19.3.453
    [94] Hicks GG, Singh N, Nashabi A, et al. (2000) Fus deficiency in mice results in defective B-lymphocyte development and activation, high levels of chromosomal instability and perinatal death. Nat Genet 24: 175-179. doi: 10.1038/72842
    [95] Kabashi E, Bercier V, Lissouba A, et al. (2011) FUS and TARDBP but not SOD1 interact in genetic models of amyotrophic lateral sclerosis. PLoS Genet 7: e1002214. doi: 10.1371/journal.pgen.1002214
    [96] Mitchell JC, McGoldrick P, Vance C, et al. (2013) Overexpression of human wild-type FUS causes progressive motor neuron degeneration in an age- and dose-dependent fashion. Acta Neuropathol 125: 273-288. doi: 10.1007/s00401-012-1043-z
    [97] Levine TP, Daniels RD, Gatta AT, et al. (2013) The product of C9orf72, a gene strongly implicated in neurodegeneration, is structurally related to DENN Rab-GEFs. Bioinformatics 29: 499-503. doi: 10.1093/bioinformatics/bts725
    [98] Farg MA, Sundaramoorthy V, Sultana JM, et al. (2014) C9ORF72, implicated in amytrophic lateral sclerosis and frontemporal dementia, regulates endosomal trafficking. Hum Mol Genet 23: 3579-3595. doi: 10.1093/hmg/ddu068
    [99] Donnelly CJ, Zhang PW, Pham JT, et al. (2013) RNA toxicity from the ALS/FTD C9ORF72 expansion is mitigated by antisense intervention. Neuron 80: 415-428. doi: 10.1016/j.neuron.2013.10.015
    [100] Therrien M, Rouleau GA, Dion PA, et al. (2013) Deletion of C9ORF72 results in motor neuron degeneration and stress sensitivity in C. elegans. PLoS One 8: 1-10. doi: 10.1371/journal.pone.0083450
    [101] Ciura S, Lattante S, Le Ber I, et al. (2013) Loss of function of C9orf72 causes motor deficits in a zebrafish model of amyotrophic lateral sclerosis. Ann Neurol 74: 180-187.
    [102] Koppers M, Blokhuis AM, Westeneng H-J, et al. (2015) C9orf72 ablation in mice does not cause motor neuron degenerateon or motor deficit. Ann Neurol 78: 426-438. doi: 10.1002/ana.24453
    [103] Ranum LPW, Cooper TA (2006) RNA-mediated neuromascular disorders. Annu Rev Neurosci 29: 259-77.
    [104] Xu Z, Poidevin M, Li X, et al. (2013) Expanded GGGGCC repeat RNA associated with amyotrophic lateral sclerosis and frontotemporal dementia causes neurodegeneration. Proc Natl Acad Sci U S A 110: 7778-7783. doi: 10.1073/pnas.1219643110
    [105] Haeusler AR, Donnelly CJ, Periz G, et al. (2014) C9orf72 nucleotide repeat structures initiate molecular cascades of disease. Nature 507: 195-200.
    [106] Zu T, Gibbens B, Doty NS, et al. (2010) Non-ATG-initiated translation directed by microsatellite expansions. Proc Natl Acad Sci U S A 108: 260-265.
    [107] Mori K, Weng S, Arzberger T, et al. (2013) The C9orf72 GGGGCC repeat is translated into aggregating dipeptide-repeat proteins in FTLD/ALS. Science 339: 1335-1339. doi: 10.1126/science.1232927
    [108] Mori K, Lammich S, Mackenzie IR, et al. (2013) hnRNP A3 bind to GGGGCC repeats and is a constituent of p62-positive/TDP-43-negative inclusions in the hippocampus of patients with C9orf72 mutations. Acta. Neuropathol 125: 413-423. doi: 10.1007/s00401-013-1088-7
    [109] Ash PEA, Bieniek KF, Gendron TF, et al. (2013) Unconventional translation of C9ORF72 GGGGCC expansion generates insoluble polypeptides specific to c9FTD/ALS. Neuron 77: 639-646. doi: 10.1016/j.neuron.2013.02.004
    [110] Zhang YJ, Jansen-West K, Xu YF, et al. (2014) Aggregation-prone c9FTD/ALS poly(GA) RAN-translated proteins cause neurotoxicity by inducing ER stress. Acta Neuropathol 128: 505-524. doi: 10.1007/s00401-014-1336-5
    [111] Mizielinska S, Grönke S, Niccoli T, et al. (2014) C9orf72 repeat expansions cause neurodegeneration in Drosophila through arginine-rich proteins. Science 345: 1192-1195. doi: 10.1126/science.1256800
    [112] Tran H, Almeida S, Moore J, et al. (2015) Differential toxicity of nuclear RNA foci versus dipeptide repeat proteins in a Drosophila model of C9ORF72 FTD/ALS. Neuron 87: 1207-1214 doi: 10.1016/j.neuron.2015.09.015
    [113] Chew J, Gendron TF, Prudencio M, et al. (2015) C9ORF72 repeat expansion in mice cause TDP-43 pathology, neuronal loss, and behavioral deficits. Science 348: 2-6.
    [114] O’Rourke JG, Bogdanik L, Muhammad AKMG, et al. (2015) C9orf72 BAC Transgenic Mice Display Typical Pathologic Features of ALS/FTD. Neuron 88: 892-901. doi: 10.1016/j.neuron.2015.10.027
    [115] Zhang K, Donnelly CJ, Haeusler AR, et al. (2015) The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature 525: 56-61. doi: 10.1038/nature14973
    [116] Freibaum BD, Lu Y, Lopez-gonzalez R, et al. (2015) GGGGCC repeat expansion in C9orf72 compromises nucleocytoplasmic transport. Nature 525: 129-133. doi: 10.1038/nature14974
    [117] Jovičić A, Mertens J, Boeynaems S, et al. (2015) Modifiers of C9orf72 dipeptide repeat toxicity connect nucleocytoplasmic transport defects to FTD/ALS. Nat Neurosci 18: 1226-1229. doi: 10.1038/nn.4085
    [118] Rezaie T, Child A, Hitchings R, et al. (2002) Adult-onset primary open-angle glaucoma caused by mutations in optineurin. Science 295: 1077-1079. doi: 10.1126/science.1066901
    [119] Zhu G, Wu CJ, Zhao Y, et al. (2007) Optineurin negatively regulates TNFα- induced NF-κB activation by competing with NEMO for ubiquitinated RIP. Curr Biol 17: 1438-1443. doi: 10.1016/j.cub.2007.07.041
    [120] Wild P, Farhan H, McEwan DG, et al. (2011) Phosphorylation of the autophagy receptor Optineurin restricts salmonella growth. Science 333: 228-233.
    [121] Sahlender DA, Roberts RC, Arden SD, et al. (2005) Optineurin links myosin VI to the Golgi complex and is involved in Golgi organization and exocytosis. J Cell Biol 169: 285-295. doi: 10.1083/jcb.200501162
    [122] Wild P, Farhan H, McEwan DG, et al. (2011) Phosphorylation of the autophagy receptor optineurin restricts Salmonella growth. Science 333: 228-233. doi: 10.1126/science.1205405
    [123] Deng HX, Bigio EH, Zhai H, et al. (2012) Differential involvement of Optineurin in amyotrophic lateral sclerosis with or without SOD1 mutations. Arch Neurol 68: 1057-1061.
    [124] Ito H, Fujita K, Nakamura M, et al. (2011) Optineurin is co-localized with FUS in basophilic inclusions of ALS with FUS mutation and in basophilic inclusion body disease. Acta Neuropathol 121: 555-557. doi: 10.1007/s00401-011-0809-z
    [125] Williams KL, Warraich ST, Yang S, et al. (2012) UBQLN2/ubiquilin 2 mutation and pathology in familial amyotrophic lateral sclerosis. Neurobiol Aging 33: 2527.e3-10.
    [126] Seok Ko H, Uehara T, Tsuruma K, et al. (2004) Ubiquilin interacts with ubiquitylated proteins and proteasome through its ubiquitin-associated and ubiquitin-like domains. FEBS Lett 566: 110-114. doi: 10.1016/j.febslet.2004.04.031
    [127] Ritson GP, Custer SK, Freibaum BD, et al. (2010) TDP-43 mediates degeneration in a novel Drosophila model of disease caused by mutations in VCP/p97. J Neurosci 30: 7729-7739. doi: 10.1523/JNEUROSCI.5894-09.2010
    [128] Ye Y, Shibata Y, Yun C, et al. (2004) A membrane protein complex mediates retro-translocation from the ER lumen into the cytosol. Nature 429: 841-847. doi: 10.1038/nature02656
    [129] Ye Y, Shibata Y, Kikkert M, et al. (2005) Recruitment of the p97 ATPase and ubiquitin ligases to the site of retrotranslocation at the endoplasmic reticulum membrane. Proc Natl Acad Sci U S A 102: 14132-14138. doi: 10.1073/pnas.0505006102
    [130] Song C, Wang Q, Li CH (2003) ATPase Activity of p97-Valosin-containing Protein (VCP). D2 mediates the major enzyme activity, and D1 contributes to the heat-induced activity. J Biol Chem 278: 3648-3655. doi: 10.1074/jbc.M208422200
    [131] Carvalho P, Stanley AM, Rapoport TA (2010) Retrotranslocation of a misfolded luminal ER protein by the ubiquitin-ligase Hrd1p. Cell 143: 579-591. doi: 10.1016/j.cell.2010.10.028
    [132] Bilican B, Serio A, Barmada SJ, et al. (2012) Mutant induced plu- ripotent stem cell lines recapitulate aspects of TDP-43 proteinopathies and reveal cell-specific vulnerability. Proc Natl Acad Sci U S A 109: 5803-5808. doi: 10.1073/pnas.1202922109
    [133] Liu X, Chen J, Liu W, et al. (2015) The fused in sarcoma protein forms cytoplasmic aggregates in motor neurons derived from integration-free induced pluripotent stem cells generated from a patient with familial amyotrophic lateral sclerosis carrying the FUS-P525L mutation. Neurogenetics 16: 223-231. doi: 10.1007/s10048-015-0448-y
    [134] Mackenzie IR, Bigio EH, Ince PG, et al. (2007) Pathological TDP-43 distinguishes sporadic amyotrophic lateral sclerosis with SOD1 mutations. Ann Neurol 61: 427-434. doi: 10.1002/ana.21147
    [135] Yamanaka K, Chun SJ, Boillee S, et al. (2008) Astrocytes as determinants of disease progression in inherited amyotrophic lateral sclerosis. Nat Neurosci 11: 251-253. doi: 10.1038/nn2047
  • This article has been cited by:

    1. Sompriya Chatterjee, Abbas Salimi, Jin Yong Lee, Insights into amyotrophic lateral sclerosis linked Pro525Arg mutation in the fused in sarcoma protein throughin silicoanalysis and molecular dynamics simulation, 2020, 0739-1102, 1, 10.1080/07391102.2020.1794967
    2. Hyun-Jun Choi, Ji Young Lee, Sun Joo Cha, Yeo Jeong Han, Ja Hoon Yoon, Hyung-Jun Kim, Kiyoung Kim, FUS-induced neurotoxicity is prevented by inhibiting GSK-3β in a Drosophila model of amyotrophic lateral sclerosis, 2022, 31, 0964-6906, 850, 10.1093/hmg/ddab290
  • Reader Comments
  • © 2016 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(8361) PDF downloads(1496) Cited by(2)

Article outline

Figures and Tables

Figures(3)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog