Published online: September 15, 2017

Review

Dysregulated molecular pathways in amyotrophic lateral sclerosis–frontotemporal dementia spectrum disorder Fen-Biao Gao*

, Sandra Almeida & Rodrigo Lopez-Gonzalez

Abstract Frontotemporal dementia (FTD), the second most common form of dementia in people under 65 years of age, is characterized by progressive atrophy of the frontal and/or temporal lobes. FTD overlaps extensively with the motor neuron disease amyotrophic lateral sclerosis (ALS), especially at the genetic level. Both FTD and ALS can be caused by many mutations in the same set of genes; the most prevalent of these mutations is a GGGGCC repeat expansion in the first intron of C9ORF72. As shown by recent intensive studies, some key cellular pathways are dysregulated in the ALSFTD spectrum disorder, including autophagy, nucleocytoplasmic transport, DNA damage repair, pre-mRNA splicing, stress granule dynamics, and others. These exciting advances reveal the complexity of the pathogenic mechanisms of FTD and ALS and suggest promising molecular targets for future therapeutic interventions in these devastating disorders. Keywords ALS; C9ORF72; FTD; FUS; TDP-43 DOI 10.15252/embj.201797568 | Received 12 June 2017 | Revised 15 July 2017 | Accepted 30 August 2017

Introduction Dementia is one of the most daunting global health challenges of the 21st century. Millions of new cases of this age-dependent condition are diagnosed each year, and more than 100 million people worldwide will be living with dementia by 2,050 (Prince et al, 2013). The cost for the social care and treatment of these patients is increasing rapidly and is estimated to reach $1 trillion next year (Wimo et al, 2017). Unfortunately, the pathogenic mechanisms underlying different forms of dementia are still poorly understood, and effective therapies have yet to be developed. Frontotemporal dementia (FTD) is the second most common form of pre-senile dementia after Alzheimer’s disease. The various forms of FTD, such as behavioral variant FTD, semantic dementia, and progressive nonfluent aphasia, are caused by focal degeneration

of the prefrontal and/or temporal cortex, resulting in changes in personality, social behaviors, or language production, as well as other cognitive impairments (Neary et al, 1998). FTD is also associated with several other neurodegenerative disorders such as progressive supranuclear palsy, corticobasal syndrome, and amyotrophic lateral sclerosis (ALS). ALS, or Lou Gehrig’s disease, is an adult-onset neurodegenerative disorder associated with progressive motor neuron loss in the brain and spinal cord, resulting in paralysis, respiratory failure, and death mostly within 3–5 years after diagnosis (Brown, 1997). Clinical dementia in ALS patients was first reported decades ago along with notable atrophy of the frontal and temporal lobes in some cases (Hudson, 1981; Horoupian et al, 1984). With better diagnosis of FTD (Neary et al, 1998) has come greater recognition of the significant clinical overlaps between ALS and FTD (Vercelletto et al, 1999; Lomen-Hoerth et al, 2002; Strong et al, 2003). The notion that ALS and FTD are related and represent two ends of a spectrum disorder is further supported by a large body of pathological and genetic evidence accumulated during the last decade or so. In 2006, a seminal discovery was made with the finding that TDP-43, encoded by the transactive response DNA binding protein (TARDBP) gene, is a major component of the pathological aggregates in both ALS and FTD brains (Arai et al, 2006; Neumann et al, 2006). TDP-43 pathology—including cytoplasmic inclusions and nuclear depletion of TDP-43—is present in more than 95% ALS cases (Ling et al, 2013). The importance of TDP-43 in ALS is further highlighted by the identification of its disease-causing mutations (Kabashi et al, 2008; Rutherford et al, 2008; Sreedharan et al, 2008; Yokoseki et al, 2008). The term frontotemporal lobar degeneration (FTLD) refers to the predominant pathological conditions in FTD patients; about half of FTLD cases have TDP-43 pathology (FTLD-TDP) (Mackenzie et al, 2010). In addition to TDP-43, genetic mutations and pathological inclusions associated with another DNA/RNA binding protein, fused in sarcoma (FUS), are found in a small percentage of ALS and FTLD cases (FTLD-FUS) (Kwiatkowski et al, 2009; Neumann et al, 2009; Vance et al, 2009). Both TDP-43 and FUS are involved in multiple aspects of RNA metabolism, and the frequent presence of either TDP-43 or FUS pathology in both ALS and FTD strongly suggests a

Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA *Corresponding author. Tel: +1 508 856 8504; E-mail: [email protected]

ª 2017 The Authors

The EMBO Journal

1

Published online: September 15, 2017

The EMBO Journal

Pathogenic mechanisms in ALS and FTD

FTD

• VCP • CHMP2B • TARDBP • FUS • UBQLN2 • SQSTM1 • C9ORF72 • CHCHD10 • OPTN • TBK1 • CCNF • TIA1

• SOD1 • hnRNP A1 • NEK1 • Others

© EMBO

• MAPT • GRN

ALS

Figure 1. Genes with genetic mutations that cause both FTD and ALS. These genes are listed chronologically by year of discovery of their involvement in FTD and ALS. Two major pathological proteins in both FTD and ALS—TDP-43 (encoded by the TARDBP gene) and FUS—are also listed here.

common pathogenic mechanism involving defective RNA metabolism (Ling et al, 2013). The overlap between ALS and FTD is most striking at the genetic level. In the year when TDP-43 was discovered as a major pathological protein in ALS and FTD (Arai et al, 2006; Neumann et al, 2006), a locus on chromosome 9p13.3–21.3 was linked to both disorders (Morita et al, 2006; Vance et al, 2006). Unlike all other ALS/FTD disease genes, the genetic mutation at this locus is a GGGGCC (G4C2) repeat expansion in the first intron of C9ORF72 (DeJesusHernandez et al, 2011; Renton et al, 2011), which turns out to be the most common genetic cause in both disorders, providing further strong evidence that they share some common pathogenic mechanisms. Remarkably, most other genes whose mutations cause FTD are also involved in ALS (Fig 1), including valosin-containing protein (VCP) (Watts et al, 2004; Johnson et al, 2010), charged multivesicular body protein 2B (CHMP2B) (Skibinski et al, 2005; Parkinson et al, 2006), ubiquilin 2 (UBQLN2) (Deng et al, 2011), sequestosome 1 (SQSTM1/P62) (Fecto et al, 2011; Rubino et al, 2012), coiled-coil-helix-coiled-coil-helix domain containing 10 (CHCHD10) (Bannwarth et al, 2014), optineurin (OPTN) (Maruyama et al, 2010; Pottier et al, 2015), TANK-binding kinase 1 (TBK1) (Cirulli et al, 2015; Freischmidt et al, 2015; Pottier et al, 2015), cyclin F (CCNF) (Williams et al, 2016), and TIA1 (Mackenzie et al, 2017). This large array of ALS/FTD genes offers exciting entry points to dissect the pathogenic mechanisms of these diseases and raises many interesting questions: How do mutations in different genes cause the same disease? What are the common molecular pathways or neural circuits affected by different genetic mutations? And how do mutations in the same gene cause different clinical phenotypes?

Molecular pathological hallmarks in C9ORF72related ALS/FTD C9ORF72 and its protein products were completely uncharacterized when G4C2 repeat expansion was first discovered in 2011. C9ORF72

2

The EMBO Journal

Fen-Biao Gao et al

mRNA has at least three variants that arise from alternative splicing and the use of different transcription start sites. Over the years, different names for these variants were used, which has caused some confusion in the field. Here, we use the latest nomenclature in the NCBI database. In variant 1 (NM_145005.6, V1) and variant 3 (NM_001256054.2, V3), G4C2 repeats are located in the first intron of C9ORF72. In contrast, transcription of variant 2 (NM_018325.4, V2) starts downstream of G4C2 repeats (Fig 2). The wildtype C9ORF72 allele mostly has fewer than 20–25 copies of these repeats, while in C9ORF72 ALS/FTD patients, there can be up to thousands (DeJesus-Hernandez et al, 2011; Renton et al, 2011; Gijselinck et al, 2012). G4C2 repeat expansion reduces the production of V2 mRNA in C9ORF72 patient cells or tissues (DeJesus-Hernandez et al, 2011; Almeida et al, 2013; Donnelly et al, 2013; Waite et al, 2014; van Blitterswijk et al, 2015). Some laboratories also reported a decrease in total level of V2 and V3 mRNAs that encode full-length C9ORF72 (DeJesus-Hernandez et al, 2011; Gijselinck et al, 2012; Belzil et al, 2013), which is probably primarily due to a decrease in V2, since V2 is expressed at a substantially higher level than V3 (and V1) in human neurons (Sareen et al, 2013; Tran et al, 2015). Decreased C9ORF72 protein levels in patient brain tissues have also been reported (Xiao et al, 2015). Another molecular pathological hallmark in C9ORF72 ALS/FTD is nuclear RNA foci (Fig 2). In many repeat-expansion diseases, expanded repeat RNAs form nuclear aggregates, which in principle can sequester important RNA binding proteins, resulting in misregulation of RNA processing (Mohan et al, 2014). Indeed, both sense and antisense RNAs transcribed from expanded C9ORF72 repeats form mostly nuclear RNA foci in different cell types, including brain neurons and primary fibroblasts from patients, as well as neurons and astrocytes derived from induced pluripotent stem cells (iPSCs) (DeJesus-Hernandez et al, 2011; Almeida et al, 2013; Donnelly et al, 2013; Gendron et al, 2013; Lagier-Tourenne et al, 2013; Lee et al, 2013; Mizielinska et al, 2013; Sareen et al, 2013; Zu et al, 2013; Lopez-Gonzalez et al, 2016). The third pathological hallmark in C9ORF72 ALS/FTD is the accumulation of dipeptide repeat (DPR) proteins, synthesized from both sense and antisense repeat transcripts as well as adjacent intronic sequences, mostly in the cytoplasm of patient neurons and glial cells (Ash et al, 2013; Gendron et al, 2013; Mori et al, 2013a,c; Zu et al, 2013; Mackenzie et al, 2013, 2015; Schludi et al, 2015; Vatsavayai et al, 2016; Fig 2). DPR proteins can be produced from overexpressed, engineered poly(A)+ mRNAs that contain expanded G4C2 repeats but without the AUG start codon, a process named as repeat-associated non-AUG (RAN) translation (Zu et al, 2011, 2013). In a recent study of mouse models of fragile X-associated tremor/ataxia syndrome, translation of expanded CGG repeats in their native molecular context commences at a near-cognate start codon in an upstream open reading frame (uORF) (Sellier et al, 2017), raising the intriguing possibility that at least some endogenous DPR proteins in C9ORF72 ALS/FTD can also be synthesized from spliced or unspliced intronic expanded G4C2 repeats through conventional ribosomal scanning for near-cognate start codons (Gao & Richter, 2017; Sellier et al, 2017). Both RNA foci and DPR proteins are present in several mouse models of C9ORF72 ALS/FTD (Chew et al, 2015; O’Rourke et al, 2015; Peters et al, 2015; Jiang et al, 2016; Liu et al, 2016). However, the toxic molecular species primarily responsible for disease

ª 2017 The Authors

Published online: September 15, 2017

Fen-Biao Gao et al

The EMBO Journal

Pathogenic mechanisms in ALS and FTD

Variant 1 NM_145005.6 Variant 2 NM_018325.4 Variant 3 NM_001256054.2

G4C2

mRNA

Repeat RNA m7G

Sense Antisense

AAAA

7

m G

AAAA

m7G

AAAA

Poly(GA) Poly(GR) Longin

Isoform b

Longin

DENN

Poly(PR)

RBP

RBP

RBP

Alpha

Isoform a

Gly

RBP

Poly(PA) Poly(GP)

Ala Pro Arg WDR41

TBK1

P C9ORF72 SMCR8 GDP

RAB

LC3-II

RNA Foci

DPR Accumulation

Phagophore © EMBO

P62

P Optineurin

Role in Autophagy

Figure 2. The C9ORF72 locus and its downstream RNA and protein products. The three C9ORF72 variants are named based on the latest information in the NCBI database. Expanded G4C2 repeat-containing intron and G2C4 repeat-containing RNA generated through an unknown mechanism can form either RNA foci or be translated into DPR proteins. Unspliced pre-mRNA may also serve as the template for DPR production (not shown). C9ORF72 isoform a contains a DENN domain and interacts with SMCR8 in autophagy.

pathogenesis remain to be fully clarified (Gendron & Petrucelli, 2017; Moens et al, 2017). Nonetheless, several major downstream pathways are commonly misregulated in both ALS and FTD caused by C9ORF72 and other genetic mutations, which will be discussed in detail below.

Autophagy Macroautophagy, the most prevalent form of autophagy, is an evolutionarily conserved, highly regulated catabolic process in which some cellular contents such as misfolded proteins and damaged organelles are targeted to lysosomes for degradation. Autophagy has been implicated in many human diseases, including different forms of neurodegeneration (Nixon, 2013; Menzies et al, 2017). This cellular process requires the formation of doublemembrane phagophores triggered by the activation of the ULK1

ª 2017 The Authors

(also known as Atg1) complex. The elongation of phagophore membranes is dependent on two ubiquitin-like conjugation systems: the covalent conjugation of Atg5 and Atg12 and lipid conjugation of the soluble form of microtubule-associated protein light chain 3 (LC3-I) that becomes the phagophore-associated form (LC3-II). Phagophore membranes seal through a poorly understood mechanism to form autophagosomes that further mature and fuse with endosomes. Mature autophagosomes then eventually fuse with lysosomes to form autolysosomes for content degradation. Cortical neurons derived from C9ORF72 iPSCs show increased sensitivity to autophagy inhibitors, which is accompanied by an elevated level of p62, a key autophagy adaptor molecule, suggesting a compromised autophagy pathway (Almeida et al, 2013). This defect could be caused in principle by partial loss of C9ORF72, the production of DPR proteins, or both. C9ORF72 is predicted to contain a DENN (differentially expressed in normal and neoplasia) domain characteristic of Rab GDP/GTP exchange

The EMBO Journal

3

Published online: September 15, 2017

The EMBO Journal

factors that activate Rab GTPases in various membrane trafficking events (Zhang et al, 2012; Levine et al, 2013). Indeed, C9ORF72 may regulate endocytosis and autophagy (Farg et al, 2014), and a flurry of recent studies showed that C9ORF72 physically binds to SMCR8 (Smith–Magenis syndrome chromosomal region candidate gene eight) (Amick et al, 2016; Sellier et al, 2016; Sullivan et al, 2016; Ugolino et al, 2016; Yang et al, 2016; Jung et al, 2017), which is another DENN domain-containing protein originally identified as a component of the autophagy interaction network (Behrends et al, 2010). Detailed molecular analyses from these studies suggested that C9ORF72 acts at different steps of the autophagy pathway. C9ORF72, WDR41, and SMCR8 form a multimeric protein complex that acts as a GDP/GTP exchange factor for Rab39b, a Rab GTPase with a role in autophagy (Sellier et al, 2016; Yang et al, 2016; Fig 2). C9ORF72 also directly or indirectly interacts with ULK1 (Sellier et al, 2016; Sullivan et al, 2016; Webster et al, 2016; Yang et al, 2016; Jung et al, 2017). The functional significance of the interaction of C9ORF72 with these autophagy regulators seems to depend on the specific cellular context. In one study, the C9ORF72/SMCR8 complex was phosphorylated by TBK1 or ULK1, leading to activation of Rab39b, which has little effect on basal autophagy but impairs autophagosome formation and causes p62+ aggregates to accumulate when autophagy is activated by mTOR inhibition (Sellier et al, 2016). A positive role for C9ORF72 and SMCR8 in autophagy induction is also supported by findings in knockout mice and patient neurons (Sullivan et al, 2016; Webster et al, 2016; Yang et al, 2016). In contrast, other studies reported that SMCR8 negatively regulates autophagy induction (Jung et al, 2016) and that loss of C9ORF72 increases autophagic flux and decreases the p62 level (Ugolino et al, 2016). The discrepancies between these studies may reflect different experimental conditions and cell types used. Further investigations are needed to clarify exactly how autophagy is misregulated in cell types that are directly relevant to C9ORF72 ALS/FTD. Numerous studies show that defects in the autophagy pathway cause neurodegeneration (Nixon, 2013; Menzies et al, 2017). However, neuronal cell loss was absent in C9orf72 knockout mice, although they developed splenomegaly and lymphadenopathy (Koppers et al, 2015; Atanasio et al, 2016; Burberry et al, 2016; Jiang et al, 2016; O’Rourke et al, 2016; Sullivan et al, 2016; Ugolino et al, 2016), suggesting that C9orf72 is not essential for basal autophagy in the nervous system. It is possible that partial loss of C9ORF72 in concert with toxic DPR proteins or other genetic mutations may result in more dramatic disease relevant phenotypes, consistent with the proposed “two-hit” mechanism in ALS/FTD (Pesiridis et al, 2011; Vance et al, 2013; Sellier et al, 2016) as well as the oligogenicity of at least some ALS cases (van Blitterswijk et al, 2012). Interestingly, under starvation conditions, C9ORF72 localizes to the surface of lysosomes, and loss of the mouse gene C9orf72 results in enlarged lysosomes (Amick et al, 2016; O’Rourke et al, 2016). Lysosomal defects are common in neurodegenerative diseases, such as FTD caused by progranulin haploinsufficiency (Kao et al, 2017). Like C9ORF72, tau binds to lysosomal membranes (Wang et al, 2009), and progranulin and its cleavage product granulin are localized inside the lysosome (Kao et al, 2017). It will be informative to further investigate how these disease proteins regulate lysosomal function at the molecular level.

4

The EMBO Journal

Pathogenic mechanisms in ALS and FTD

Fen-Biao Gao et al

Many ALS/FTD disease proteins besides C9ORF72 participate directly in the autophagy pathway, suggesting a major pathogenic mechanism in these disorders (Almeida & Gao, 2016). Some disease proteins, such as p62 and optineurin, function as autophagic adaptor proteins. In addition to its function in several signaling events, p62 binds to many substrates for selective autophagy as well to LC3-II on phagophore membranes through its LC3-interacting region (LIR) (Moscat et al, 2016). Some ALS-associated mutations seem to induce a loss of function in a zebrafish model (Lattante et al, 2015), and the L341V-mutant LIR has a greatly reduced ability to bind to LC3-II (Goode et al, 2016a). Other ALS/FTD-associated mutations compromise p62’s ability to activate the signaling mediated by the transcription factor Nrf2, which is critical for cell defense against oxidative stress, especially in neurons (Goode et al, 2016b). Like p62, optineurin interacts with polyubiquitinated cargo through an LIR domain, an ubiquitin-binding domain, and a C-terminal coiledcoil domain. Loss of optineurin reduces the clearance of protein aggregates or defective mitochondria (Korac et al, 2013; Wong & Holzbaur, 2014). ALS/FTD-associated mutations in the ubiquitinbinding domain either reduce the normal function of optineurin by affecting its ability to interact with LC3 or act in a dominantnegative manner to compromise autophagic activity (Wong & Holzbaur, 2014; Shen et al, 2015). Disease mutations in optineurin such as E696K also disrupt the formation of an optineurin/TBK1 complex (Li et al, 2016). Functionally, phosphorylation of optineurin by TBK1 promotes the binding of optineurin to its cargoes such as damaged mitochondria (Heo et al, 2015; Richter et al, 2016). TBK1 also directly binds to and phosphorylates p62 (Heo et al, 2015; Matsumoto et al, 2015) and the C9ORF72/SMCR8 complex (Sellier et al, 2016). Since many disease-associated mutations in p62, optineurin, C9ORF72, and TBK1 cause partial loss of function in these genes, suboptimal autophagy induction seems to be a key pathogenic mechanism in ALS/FTD. This mechanism seems to be especially important in microglia, which are important for synaptic pruning and debris clearance throughout the nervous system (Hong et al, 2016). For instance, progranulin is highly expressed in activated microglia and suppresses aberrant microglial activation during aging (Lui et al, 2016). Moreover, C9orf72-null mice also exhibit an altered immune response in microglia (O’Rourke et al, 2016), suggesting novel molecular links between autophagy and neuroinflammation, in addition to well-documented role for TBK1 (Oakes et al, 2017). The importance of autophagy in ALS/FTD is further supported by rare disease-causing mutations in other genes that also regulate this pathway. A splicing site mutation in CHMP2B, a subunit of the endosomal sorting complexes required for transport (ESCRT-III), causes FTD linked to chromosome 3 (FTD-3) in a large Danish family (Skibinski et al, 2005). Other CHMP2B mutations have also been implicated in ALS (Parkinson et al, 2006; Cox et al, 2010). Expression of FTD-3-associated mutant protein CHMP2BIntron5, which lacks the C-terminal 36 amino acids, causes autophagosomes and multilamellar bodies to accumulation abnormally (Filimonenko et al, 2007; Lee et al, 2007). This phenotype is likely caused by dominant-negative effect of CHMP2BIntron5, since CHMP2BIntron5 binds to other ESCRT-III subunits such as Snf-7/CHMP4B more avidly than to wildtype CHMP2B (Lee et al, 2007); however, the gene itself is not required for neuronal survival or mouse viability (Lee et al, 2007; Ghazi-Noori et al, 2012). Moreover, autophagy

ª 2017 The Authors

Published online: September 15, 2017

Fen-Biao Gao et al

Pathogenic mechanisms in ALS and FTD

requires ESCRT-III, which likely regulates the maturation of autophagosomes (Filimonenko et al, 2007; Lee et al, 2007; Rusten et al, 2007). Indeed, a strong genetic modifier of CHMP2BIntron5 toxicity, syntaxin 13, is highly localized on multilamellar bodies induced by dysfunctional ESCRT-III, further suggesting a role for ESCRT-III in autophagosome maturation (Lu et al, 2013). Mutant CHMP2B also causes lysosomal pathology (Clayton et al, 2015), which may indirectly affect the autophagy pathway. The importance of compromised autophagy is further supported by the finding that transgenic mice expressing CHMP2BIntron5 have several hallmarks of FTD-3, such as p62- and ubiquitin-positive aggregates and social behavioral deficits (Ghazi-Noori et al, 2012; Gascon et al, 2014; Vernay et al, 2016; Clayton et al, 2017). VCP, a multifunctional AAA-type ATPase, has a key role not only in ubiquitin-dependent degradation by the proteasome but also in autophagy, endosomal sorting, intracellular signaling pathways such as the NFjB pathway, and other cellular processes (Meyer et al, 2012). For instance, cells expressing FTD-associated mutant VCP are more sensitive to proteasome inhibition and accumulate ubiquitinated proteins (Ju et al, 2008). Similarly, several mutant ubiquilin-2 proteins associated with ALS fail to deliver ubiquitinated cargoes, including aggregated proteins, to proteasomes (Chang & Monteiro, 2015; Hjerpe et al, 2016; Le et al, 2016). Ubiquilin 1 and ubiquilin 2 also have a function in autophagy (N’Diaye et al, 2009; Rothenberg et al, 2010), but it is unclear how disease-related mutations in ubiquilin 2 affect this pathway. Loss of VCP or expression of FTD-associated mutant VCP results in the accumulation of immature autophagosomes, TDP-43 pathology, and reduced mTOR activity (Ju et al, 2009; Tresse et al, 2010; Ching et al, 2013). VCP is also required for autophagic clearance of damaged mitochondria (Kim et al, 2013a), stress granules (Buchan et al, 2013), and ruptured lysosomes (Papadopoulos et al, 2017). This function, too, is compromised by mutations found in ALS and FTD associated with inclusion body myopathy. Taken together, studies on different disease genes strongly suggest that defects in different steps of the autophagy pathway are major contributors to ALS/FTD pathogenesis.

Nucleocytoplasmic transport Compromised autophagy activity may lead to the formation and accumulation of large protein aggregates or inclusions. Under some circumstances, these structures can be neuroprotective, as in the case of intranuclear inclusions in Huntington’s disease (HD) (Saudou et al, 1998; Arrasate et al, 2004). On the other hand, abnormal protein aggregates adversely affect the survival of neuronal and nonneuronal cells through multiple pathways. For instance, cytoplasmic, but not nuclear, toxic aggregates, sequester and mislocalize proteins involved in nucleocytoplasmic transport (Woerner et al, 2016). Active transport of proteins and RNAs through nuclear pore complexes (NPCs) is an essential cellular process in which the small GTPase Ran regulates the interaction between transport receptors, such as the importin family of proteins, and cargo molecules (Gorlich & Kutay, 1999). Each NPC is a large macromolecular machine composed of hundreds of nuclear pore proteins belonging to about 30 different nucleoporins (NUPs) (Beck & Hurt, 2017). Some NUPs, such as the NUP107/160

ª 2017 The Authors

The EMBO Journal

subcomplex, have an extremely long half-life in postmitotic cells and are therefore susceptible to age-related deterioration, which may result in loss of nuclear integrity (D’Angelo et al, 2009; Savas et al, 2012; Toyama et al, 2013). Thus, abnormal nucleocytoplasmic transport likely has a major role in various age-dependent neurodegenerative diseases. In one of the earliest studies suggesting a potential defect in nucleocytoplasmic transport in ALS, importin a and b family proteins were mislocalized in a transgenic mouse model expressing mutant SOD1(G93A) (Zhang et al, 2006; Fig 3). Subsequent immunostaining analysis of sporadic and familial ALS cases with SOD1 mutations suggested that some components of NPCs localize discontinuously on the nuclear membrane (Kinoshita et al, 2009). Compromise of the nuclear import pathway may contribute to the cytoplasmic accumulation of TDP-43 or FUS in patients with ALS, FTLD-TDP-43, or FTLD-FUS. For example, in cultured cells, general blockage of nuclear import (Winton et al, 2008) or knockdown of some nuclear import factors such as karyopherin-b1 and transportin 1 (Dormann et al, 2010; Nishimura et al, 2010) causes TDP-43 and FUS to accumulate in the cytoplasm. Moreover, some nucleocytoplasmic transport proteins such as transportin 1 and NPC components show decreased expression or an abnormal subcellular distribution in postmortem CNS tissues of some ALS and FTD patients (Nishimura et al, 2010; Neumann et al, 2012; Takeuchi et al, 2013; Troakes et al, 2013; Zhang et al, 2015; Shang et al, 2017). Loss of normal TDP-43 function reduces the expression of both Ran in FTD patient cells with GRN mutations (Ward et al, 2014) and Ran-binding protein 1 in SH-SY5Y cells (Stalekar et al, 2015), suggesting a potential molecular feedback loop that may in part account for disease progression. Further underscoring the importance of nucleocytoplasmic transport in ALS, an adultonset form of ALS is associated with partial loss of GLE1, an important mediator of RNA export at the nuclear pore (Kaneb et al, 2015). The molecular mechanisms underlying the involvement of nucleocytoplasmic transport in ALS/FTD pathogenesis are further elucidated by recent studies on C9ORF72 repeat expansions (Fig 3). In C9ORF72 iPSC-derived neurons, the nucleocytoplasmic Ran gradient is decreased, and nuclear import of proteins and nuclear export of RNAs are compromised (Freibaum et al, 2015; Zhang et al, 2015). mRNAs also accumulate in the nucleus in cell lines expressing expanded G4C2 repeats (Rossi et al, 2015). To explore the underlying mechanisms, genetic screens in simple model organisms such as fruit flies and yeast were carried out independently by several collaborative teams (Freibaum et al, 2015; Jovicic et al, 2015; Zhang et al, 2015; Boeynaems et al, 2016). In a fly model in which 30 copies of G4C2 repeats with a restriction enzyme site in the middle are expressed in the 50 UTR of the reporter gene GFP (Xu et al, 2013), G4C2 repeat toxicity was suppressed by increased activity of Ran GTPase-activating protein (RanGAP), which stimulates the GTPase activity of Ran and positively regulates nucleocytoplasmic transport (Zhang et al, 2015). Moreover, nuclear import was partially inhibited, and other molecular manipulations of nucleocytoplasmic transport either enhanced or suppressed (G4C2)30 toxicity in the fly (Zhang et al, 2015). Consistent with these findings, many genetic modifiers of the rough eye phenotype that encode key proteins involved in the nucleocytoplasmic transport of both proteins and RNAs, such as

The EMBO Journal

5

Published online: September 15, 2017

The EMBO Journal

Pathogenic mechanisms in ALS and FTD

Fen-Biao Gao et al

Cargo Poly(PR)

Importin

Importin

mSOD1

Importin

Importin TDP-43 GDP

Cargo

Ran

GTP GTP

mHTT

Pi

Ran

Ran

Importin Nucleus

Gle1

RanGAP1 mHTT Nup62

Poly(GA)

THOC2

THOC2

RanGAP1

TDP-43

RNAs

RanGAP1

THOC2 RNA

Cytoplasm

THOC2 THOC2

© EMBO

HR23

G4C2 Repeats

mHTT

Poly(PR) RNA

Gly

Ala

Pro

Arg

Figure 3. Nucleocytoplasmic transport defects in different neurodegenerative diseases. In C9ORF72-related FTD/ALS, expanded G4C2 repeats bind to RanGAP and disrupt the Ran gradient; poly(PR) interacts with the FG repeats of nuclear pore proteins; poly(GR) binds to importin; and poly(GA) aggregates sequester RanGAP and HR23. Cytoplasmic and nuclear aggregates formed by TDP-43 or mutant huntingtin (mHTT) sequester some NPC components and factors important for nucleocytoplasmic transport.

Nup50, Ran, and Gle1, were identified in an unbiased genetic screen in a fly model where (G4C2)58 was expressed in the context of poly(A)+ mRNA together with the GFP coding region but without the AUG start codon (Freibaum et al, 2015). The defects in nucleocytoplasmic transport in C9ORF72-related ALS/FTD are likely caused by both toxic repeat RNAs and DPR proteins. RanGAP1 directly binds to G4C2 repeat RNA in vitro, and colocalizes with G4C2 RNA foci in C9ORF72 patient cells (Zhang et al, 2015). The notion that RanGAP1 is a novel RNA binding protein is also supported by a recent eCLIP analysis that revealed a preferential binding of the protein to the CGGCGG motif (Brannan et al, 2016). Thus, G4C2 repeat RNA might compromise nucleocytoplasmic transport by interacting physically with RanGAP1. However, since poly(GR) expression alone also causes severe retinal degeneration in the fly (Mizielinska et al, 2014; Wen et al, 2014; Yang et al, 2015), poly(GR) proteins that are readily detectable in (G4C2)58 flies may be partly responsible for the observed repeat toxicity as well (Freibaum et al, 2015). Indeed, unbiased genetic screens in yeast identified many modifiers that implicate nucleocytoplasmic transport as a key target of poly(GR) and poly(PR) toxicity (Jovicic et al, 2015). A subsequent targeted RNAi screen in Drosophila confirmed that importins, NUPs, and many other

6

The EMBO Journal

nucleocytoplasmic transport proteins strongly modify the toxicity of arginine-rich DPRs (Boeynaems et al, 2016). In addition to these genetic studies in model organisms, interactome analysis revealed that some importins, NUPs, and the lamin B receptor bind directly to poly(GR) and poly(PR) proteins (Lee et al, 2016; Lopez-Gonzalez et al, 2016; Shi et al, 2017). In particular, poly(PR) binds to the central channel of the nuclear pore and promotes polymerization of the phenylalanine:glycine domain of NUPs, thereby reducing trafficking through the pore (Shi et al, 2017). These findings point to a specific molecular mechanism by which arginine-rich DPR proteins compromise nuclear pore function and nucleocytoplasmic transport. Poly(GA) aggregates can also sequester nucleocytoplasmic transport proteins such as HR23 and induce an abnormal distribution of RanGAP1 in mice, and overexpression of HR23 or importins or NUPs in cultured cells rescues poly(GA) toxicity (Zhang et al, 2016; Khosravi et al, 2017). Moreover, transport factors are partially sequestered and nucleocytoplasmic transport is disrupted in multiple iPSC and animal models of HD and in HD patient brains (GassetRosa et al, 2017; Grima et al, 2017), further supporting the notion that defects in this cellular pathway are common in different neurodegenerative diseases (Fig 3).

ª 2017 The Authors

Published online: September 15, 2017

Fen-Biao Gao et al

Pathogenic mechanisms in ALS and FTD

DNA damage The integrity of genomic DNA in mammalian postmitotic neurons needs to be maintained without new replication for up to decades. DNA is constantly being damaged in many ways. In neurons, a major cause of such damage is endogenous reactive oxygen species (ROS). The production of ROS from mitochondrial respiration and other metabolic processes continuously generates numerous oxidative base modifications and changes in the sugar phosphate backbone of DNA, leading to single-strand breaks (SSBs) and, less frequently, to presumably more deleterious DNA double-strand breaks (DSBs) (Madabhushi et al, 2014). During gene transcription, DNA damage such as spontaneous deamination of dC and dU on the unpaired single-stranded DNA can also lead to DSBs (SkourtiStathaki & Proudfoot, 2014). Even normal neuronal activity increases neuronal DSBs, which is a physiological event, but may also contribute to neurodegeneration under pathological conditions (Suberbielle et al, 2013; Madabhushi et al, 2015). Mammalian neurons and other cell types have evolved sophisticated DNA damage response and repair mechanisms. In postmitotic cells such as neurons, DSBs are repaired primarily by the nonhomologous end-joining pathway, in which DSB sites are recognized and bound by the Ku70-Ku80 complex, which activates DNA-dependent protein kinase (DNA-PK) and recruits a host of factors, including another kinase, ataxia telangiectasia mutated (ATM), to complete the repair (Rulten & Grundy, 2017). ATM phosphorylates multiple proteins involved in the repair pathway, among them the histone H2AX, a widely used DNA damage marker, and p53, whose stabilization may lead to apoptotic cell death (Sancar et al, 2004). Defects in these DNA repair pathways contribute to many neurodegenerative diseases including ALS and FTD (Madabhushi et al, 2014), as highlighted by the involvement of FUS, TDP-43, and DPR proteins in DNA damage and repair. This notion is further strengthened by the identification of NEK1, a serine/threonine protein kinase involved in DNA damage repair and other cellular processes, and its interactor C21ORF2, as risk factors for ALS (Brenner et al, 2016; Kenna et al, 2016; van Rheenen et al, 2016). A well-studied example in which ALS/FTD disease mutations directly affect the DNA damage response is FUS. FUS participates in the formation of a D-loop during DNA DSB repair, suggesting a potential role in genomic stability (Baechtold et al, 1999). Indeed, FUS-deficient fibroblasts from knockout mice have increased sensitivity to ionizing irradiation along with chromosome abnormalities, such as chromosome breakage, centromeric fusion, and aneuploidy (Hicks et al, 2000; Kuroda et al, 2000). FUS has an important and specific role in DNA damage response. First, FUS is recruited within seconds to the site of laser-induced oxidative DNA damage and DSBs (Mastrocola et al, 2013; Wang et al, 2013; Britton et al, 2014; Patel et al, 2015). Second, FUS recruitment is dependent on the interaction between an arginine/glycine-rich domain of FUS and poly(ADP-ribose) polymerase, whose polymeric product is required to recruit many other DNA damage-response proteins, including FUS itself (Mastrocola et al, 2013; Rulten et al, 2014). Third, immobilization of FUS to chromatin can induce the formation cH2AX foci, and thus, FUS may be an early component of the DNA repair machinery (Wang et al, 2013). Fourth, FUS directly interacts with HDAC1, another critical component of the DNA damage response pathway, and this interaction is enhanced by the induction of DNA

ª 2017 The Authors

The EMBO Journal

damage (Wang et al, 2013; Qiu et al, 2014). Finally, loss of FUS reduces both HDAC1 accumulation and DNA repair activity at DSBs (Mastrocola et al, 2013; Wang et al, 2013) and increases transcription-associated DNA damage (Hill et al, 2016). Consistent with a key role for FUS in DNA damage repair, the FUS mutants R224C, R514S, H517Q, and R521C show various degrees of deficiency in DNA DSB repair (Wang et al, 2013). Accordingly, the accumulation of several proteins in the DNA repair machinery at DSB sites was reduced in cells expressing some of these mutants (Wang et al, 2013). In particular, FUS-R521C interferes with the normal interaction between FUS and HDAC1 (Wang et al, 2013; Qiu et al, 2014), and FUS-R521C transgenic mice have greatly increased DNA damage in both cortex and spinal cord (Qiu et al, 2014). Although a direct link between elevated DNA damage and eventual neuronal cell death has not been firmly established, a similar phenotype has been observed in neurons from ALS/FTD patients. For instance, iPSC-derived motor neurons of an ALS patient with a frameshift FUS mutation had an age-dependent increase in DNA damage (Higelin et al, 2016), as do motor cortex neurons in ALS patients with FUS-R521C and P525L mutations (Wang et al, 2013) and brain tissues of FTLD-FUS cases where nuclear function of FUS may be reduced (Deng et al, 2014). Phosphorylation on the N-terminal domain of FUS by DNA-PKs causes its translocation into the cytoplasm after DNA damage, suggesting that DNA damage can be the trigger that leads to mislocalization of FUS (Deng et al, 2014). Thus, compromised DNA repair caused by mutant FUS is a major potential pathogenic mechanism in ALS/FTD with FUS mutations or FUS aggregates in the cytoplasm. In contrast to FUS, the potential link between TDP-43 and DNA damage is surprisingly understudied. Although nuclear depletion of TDP-43 is a hallmark of ALS and FTD, only recently have researchers begun to investigate whether loss of TDP-43 normal function increases DNA damage. Like FUS, TDP-43 colocalizes with active RNA polymerase II and the DNA damage repair protein BRCA1 at the sites of transcription-associated DNA damage in U2OS cells (Hill et al, 2016). Moreover, knockdown of TDP-43 increases sensitivity to a-amanitin, a transcription-arresting agent (Hill et al, 2016). In support of a link between TDP-43 pathology and genomic instability, overexpression of human TDP-43 (hTDP-43) in a fly model resulted in nuclear clearance of the protein, derepression of retrotransposable elements, and cell death from increased DNA damage (Krug et al, 2017). The molecular details concerning how TDP-43 contributes to the prevention or repair of transcriptionassociated or other forms of DNA remain to be further investigated. Recently, an age-dependent increase in DNA damage and oxidative stress was reported in iPSC-derived motor neurons of patients with C9ORF72 repeat expansions (Lopez-Gonzalez et al, 2016). DNA damage is also increased in spinal cord neurons of C9ORF72 ALS patients (Farg et al, 2017; Walker et al, 2017). Expression of poly(GR) in iPSC-derived control neurons or neuronal cell lines is sufficient to induce DNA damage (Lopez-Gonzalez et al, 2016; Farg et al, 2017). In addition, reduction in oxidative stress by treatment with antioxidants partially rescues DNA damage both in motor neurons from C9ORF72 carriers and in neurons expressing (GR)80, Thus, oxidative stress may be partly responsible for increased DNA damage (Lopez-Gonzalez et al, 2016). In a Drosophila model of intronic G4C2 repeat expansion, neurodegeneration is not observed but proteins with oxidoreductase activities are enriched among a

The EMBO Journal

7

Published online: September 15, 2017

The EMBO Journal

small number of differentially expressed genes (Tran et al, 2015), suggesting that increased oxidative stress might be an early pathogenic event. It is not known whether other molecular pathways also contribute to increased DNA damage in C9ORF72 ALS/FTD.

Pre-mRNA splicing Intron removal from pre-mRNAs is highly regulated and requires the specific interactions of more than 100 proteins with cis elements such as the 50 splice site, the branch point, and the 30 splice site, as well as other regulatory elements. For the majority of pre-mRNAs, spliceosome assembly starts with the recruitment of U1 and U2 small nuclear ribonucleoproteins (snRNPs) and then other complexes. Recently, cryo-electron microscopy structural analyses provided further mechanistic insight into the workings of this highly sophisticated macromolecular machine (e.g., Rauhut et al, 2016; Yan et al, 2017). During pre-mRNA splicing, many other RNA binding proteins (RBPs) such as serine/arginine (SR) and heterogeneous nuclear RNP (hnRNP) family proteins help define the exon/intron boundaries and thereby regulate alternative splicing in a developmental stage- and cell type-specific manner. The importance of regulated pre-mRNA splicing is further underscored by widespread changes in alternative splicing and many genetic mutations in either cis elements or trans factors in various human diseases (Singh & Cooper, 2012; Chabot & Shkreta, 2016). In ALS patients with C9ORF72 repeat expansion, alternative splicing in the cerebellum and frontal cortex was extensively misregulated and more frequent than in sporadic cases (Prudencio et al, 2015). Splicing defects have also been found in lymphoblastoid cell lines derived from some C9ORF72-ALS patients (Cooper-Knock et al, 2015). Interestingly, alternative splicing events in such patients are eightfold to ninefold more frequent in the cerebellum than in the frontal cortex (Prudencio et al, 2015), which is consistent with the more pronounced DPR pathology in the cerebellum of C9ORF72 patients (Mori et al, 2013; Ash et al, 2013; Mackenzie et al, 2015). In light of these findings, it is worth highlighting recent evidence that cerebellum is also involved in cognitive processing (Wagner et al, 2017). The mis-splicing described above may be explained in part by the finding that U2 snRNP proteins are major interactors of poly (GR) and poly(PR) in nuclear extracts and that these DPR proteins block spliceosome assembly and splicing in an in vitro assay (Yin et al, 2017). These proteins are also present in the interactomes of poly(GR) and poly(PR) identified by others (Lee et al, 2016; Lin et al, 2016; Lopez-Gonzalez et al, 2016; Boeynaems et al, 2017). Moreover, some components of U2 snRNP, which normally localize to the nucleus, are mislocalized in the cytoplasm of motor neurons derived from C9ORF72 iPSCs, and nearly 50% of mis-spliced exons identified in C9ORF72 brains by Prudencio et al (2015) are U2 snRNP-dependent (Yin et al, 2017). Thus, U2 snRNP may be a major target of the toxicity of arginine-rich DPR proteins. In C9ORF72-related ALS/FTD, many proteins can bind to G4C2 repeat RNAs in vitro, and some are reported to colocalize with RNA foci in some patient cells (Almeida et al, 2013; Donnelly et al, 2013; Lee et al, 2013; Mori et al, 2013b; Reddy et al, 2013; Sareen et al, 2013; Xu et al, 2013; Cooper-Knock et al, 2014; Haeusler et al, 2014; Rossi et al, 2015; Conlon et al, 2016). Thus, sequestration of

8

The EMBO Journal

Pathogenic mechanisms in ALS and FTD

Fen-Biao Gao et al

these proteins may alter pre-mRNA splicing. On the other hand, abundant sense RNA foci produced in a Drosophila model of intronic G4C2 repeat expansion do not induce neurodegeneration (Tran et al, 2015). Moreover, in C9ORF72 patients, sense foci are not associated with any clinicopathological features, and the number of cells with antisense foci correlates inversely with the age of disease onset, suggesting a potential beneficial role (DeJesusHernandez et al, 2017). Interestingly, expanded repeat RNAs alone can undergo gelation in vitro without other proteins through a process called liquid–liquid phase separation, which may be also responsible for RNA foci formation in human cells (Jain & Vale, 2017). Thus, it remains to be determined whether enough of any protein can be sequestered by RNA foci to affect splicing in a detrimental way in C9ORF72 patients. Long before TDP-43 was identified as an FTD/ALS pathological protein (Neumann et al, 2006) and disease-causing FUS mutations were identified in ALS and FTD (Kwiatkowski et al, 2009; Vance et al, 2009), these proteins were known to have key roles in premRNA splicing (e.g., Yang et al, 1998; Buratti et al, 2001). Indeed, TDP-43 binds to thousands of different RNAs in mammalian cell lines and the brain, from microRNAs to long introns (Polymenidou et al, 2011; Sephton et al, 2011; Tollervey et al, 2011; Xiao et al, 2011; Colombrita et al, 2012; Li et al, 2013a; Narayanan et al, 2013; Fan et al, 2014). Binding of TDP-43 to very long introns (> 100 kb) affects the stability of these pre-mRNA targets, and binding to exon– intron junctions and some intronic sequences regulates alternative splicing (Polymenidou et al, 2011; Tollervey et al, 2011; Passoni et al, 2012). This regulation is partly mediated by UG-rich sequences that interact with both RNA recognition motifs of TDP-43 (Passoni et al, 2012; Lukavsky et al, 2013). Similarly, FUS binds along the whole length of thousands of nascent RNAs and regulates alternative splicing of RNAs that are mostly distinct from those dependent on TDP-43 (Lagier-Tourenne et al, 2012; Rogelj et al, 2012). Loss of TDP-43 results in aberrant mRNAs containing cryptic exons that often lead to premature stop codons and nonsensemediated decay (Ling et al, 2015). This mode of regulation targets a large number of transcripts, and many of these nonconserved cryptic exons are cell type specific (Tan et al, 2016; Jeong et al, 2017) and may occur in Alzheimer’s brains in addition to ALS/FTD cases (Sun et al, 2017). Since the majority of ALS and FTD cases show TDP-43 pathology, including depletion of TDP-43 in the nucleus, loss of normal TDP-43 function in splicing is likely to be a key pathogenic factor in disease progression. On the other hand, mutant TDP-43 can also affect splicing in a gain-of-toxic-function manner. For instance, mutant TDP-43(Q331K) expressed in vivo can either enhance or impair splicing in an RNA target-dependent manner and cause motor neuron disease without loss of nuclear TDP-43 (Arnold et al, 2013). Mutant TDP-43(M337V) also causes mislocalization of the splicing factor PSF and NeuN in the cytosol and misregulation of PSF/NeuN-dependent splicing, which is essential for neuronal function (Wang et al, 2015). Similarly, FUS interacts with U1, U11, and U12 snRNPs, which are mislocalized to the cytosol by mutant FUS (Sun et al, 2015; Yu & Reed, 2015; Yu et al, 2015; Reber et al, 2016). Moreover, both loss of function and gain of toxic function in hnRNP A2/B1 likely contribute to defects in alternative splicing in patient fibroblasts and iPSC-derived motor neurons harboring the ALS-associated mutation D290V (Martinez et al, 2016). These

ª 2017 The Authors

Published online: September 15, 2017

Fen-Biao Gao et al

The EMBO Journal

Pathogenic mechanisms in ALS and FTD

RBPs with LCD RRM

RRM

Liquid–Liquid Phase Separation

LCD

[RNA] [Salt] Temperature pH

Gly Pro Arg Poly(GR) Poly(PR)

…GGGGGGNNGGGGGYGGGGGNYGGGNGGGGGY… Examples

• FUS • TDP-43 • hnRNP A1 • hnRNP A2B1

Dispersed proteins

• TIA1 • TIAR • G3BP1

Liquid droplets

TDP-43 toxicity i Continuous stress (e.g. mutation) Translation

Irreversible aggregates

PERK elF2α TIA1

AAA

Mutant FUS

FUS

LCD LCD

AAAA PABP

7 mG

AAA

A

AA

AA

A

A

AA

AAAA AA AA A AA

ATX-2 Mutant Ataxin-2

Individual Components

AA

A

m 7G 7G

m

© EMBO

m7G

OR

7 mG

AA AA

G3BP1

AA

AA

C

m

FUS

7 AAA m G A m7G m7 AAA G A AAA A AAAA m 7G

m 7G

A

7G

AAA A

AAAA

PUM 2

AAAA PABP

AA AA

mG 40S LCD

7

G

mG m 7G

7 mG

TDP-43

RBP

7

7 m G

LCD

A A AA A A AA

7G 7 m mG

m 7G

Poly(PR)

e or

AA

AA

LCD

“stalled”

AA

7 mG

AA

h

A

LCD

P

A

m7G

m 7G

40S

elF2α

AAAA RBP PABP

S

LCD

FMRP

m7G

l el

m7

Poly(GR)

Stress Granule (SG)

Figure 4. The roles of different FTD/ALS disease proteins in SG formation. Upper left: An artificial sequence is presented to illustrate the amino acid composition of LCDs. Upper right: A schematic representation of liquid–liquid phase separation in vitro that can be influenced by salt concentrations or RNA concentrations or genetic mutations. Center: TDP-43 aggregates or inhibition of general translation by arginine-rich DPR proteins can lead to elF2a phosphorylation which promotes SG formation. Mutant proteins in FTD/ALS such as FUS or ataxin-2 promote the formation of irreversible aggregates and thereby prevent the dissociation of SGs. Lower left: A schematic representation of some individual components of SGs. LCD: LCD-containing RBPs. Lower right: A schematic representation of an SG with a densely packed core.

results underscore the important role of misregulation of splicing by different disease proteins in ALS/FTD.

Stress granules In addition to multifunctional roles in the cellular processes mentioned above, several disease proteins in ALS/FTD are also associated with stress granules (SGs). SGs are non-membranebound, cytoplasmic ribonucleoprotein (RNP) granules composed of mRNAs, translation initiation factors, 40S ribosomes, and many other RBPs (Panas et al, 2016; Protter & Parker, 2016). SGs of up to a few micrometers can be induced by various cellular stresses that inhibit translation initiation, such as oxidative stress, glucose starvation, mitochondrial dysfunction, and viral infection. These cytoplasmic structures can either disassemble upon release of cellular stress

ª 2017 The Authors

or be degraded by the autophagy pathway. As a stress response mechanism, SG formation is likely to be beneficial under certain circumstances. On the other hand, since SGs may be mechanistically linked to aggregate formation in several neurodegenerative diseases, dysregulated formation or clearance of SGs may be a key pathogenic mechanism (Wolozin, 2012; Li et al, 2013b; Ramaswami et al, 2013). The recent identification of TIA1 mutations in ALS/FTD that alter SG dynamics further highlights the importance of this pathway in neurodegeneration (Mackenzie et al, 2017). One of the best-illustrated examples of the potential link between SGs and aggregate formation is FUS. In response to oxidative stress or heat shock treatment, endogenous FUS is targeted to SGs in HeLa cells (Andersson et al, 2008). Some ALSassociated mutant FUS proteins (such as R521G) show increased cytoplasmic localization and induce SG formation with or without further stress; the increase is probably dependent on the level of

The EMBO Journal

9

Published online: September 15, 2017

The EMBO Journal

ectopic expression (Bosco et al, 2010; Gal et al, 2011; Sun et al, 2011). Indeed, in fibroblasts or iPSC-derived motor neurons containing endogenous ALS-associated FUS mutations, FUSpositive SGs accumulate only after stress treatment or neuronal aging (Japtok et al, 2015; Lenzi et al, 2015; Lim et al, 2016). The recruitment of endogenous or mutant FUS to SGs and its interaction with other SG components are dependent on RNA (Andersson et al, 2008; Bentmann et al, 2012; Daigle et al, 2013; Shelkovnikova et al, 2013; Vance et al, 2013). Another determinant of SG formation is the prionlike domain located at the N-terminus of FUS (King et al, 2012), a low-complexity domain (LCD) also present in many RBPs and other proteins that mediates liquid–liquid phase separation, a process thought to underlie the formation of many membraneless organelles such as SGs (Burke et al, 2015; Lin et al, 2015; Molliex et al, 2015; Murakami et al, 2015; Patel et al, 2015; Fig 4). ALS/FTD-associated mutations in the LC and non-LC domains of FUS further promote the transition of liquid droplets into fibrillar aggregates in vitro and therefore may prevent the proper dissociation of FUS-containing SGs in living cells (Murakami et al, 2015; Patel et al, 2015). These findings provide a plausible molecular mechanism underlying RBP aggregates in ALS/FTD. TDP-43 also associates with SGs through an interaction with the core SG protein TIA-1, and some but not other disease-related mutations in TDP-43 enhance SG formation (Freibaum et al, 2010; Liu-Yesucevitz et al, 2010; Dewey et al, 2011; McDonald et al, 2011; Bentmann et al, 2012). Similarly, disease mutations in hnRNP A2 and hnRNP A1 also increase SG formation (Kim et al, 2013b). Early observations that some TDP-43-positive inclusions in patient tissues may contain SG markers support the notion that dysregulated SG dynamics contribute to TDP-43 proteinopathies (Liu-Yesucevitz et al, 2010; Bentmann et al, 2012). Under certain experimental conditions, blocking SG assembly through G3BP knockdown increases cell death under stress, indicating a beneficial role for SG formation (Aulas et al, 2012). In contrast, many genes that affect SG formation are strong modifiers of TDP-43 toxicity in yeast and Drosophila (Kim et al, 2014). In particular, TDP-43 toxicity increases the phosphorylation of elF2a in flies, and TDP-43 toxicity is greatly mitigated by genetic reduction in PEK, which phosphorylates elF2a and is the fly homolog of mammalian PERK, or by pharmacological treatment of flies or mammalian neurons with the PERK inhibitor GSK2606414 (Kim et al, 2014). Because reduction in PERK activity and elF2a phosphorylation blocks SG formation, these results imply that prolonged SG accumulation is detrimental and that partial inhibition of this process may have therapeutic values. Consistent with this notion, either genetic reduction or antisense oligonucleotide (ASO) knockdown of the SG component ataxin-2 in the central nervous system increased the survival of TDP-43 transgenic mice (Becker et al, 2017). Recent studies have implicated abnormal SG dynamics in C9ORF72-related ALS/FTD. Arginine-rich DPR proteins suppress global translation and thus may indirectly trigger SG formation, likely reflecting their preferential direct physical interaction with ribosomal proteins (Wen et al, 2014; Kanekura et al, 2016; LopezGonzalez et al, 2016). These DPR proteins are also associated with many other RBPs (Lee et al, 2016; Lin et al, 2016; LopezGonzalez et al, 2016; Boeynaems et al, 2017; Yin et al, 2017),

10

The EMBO Journal

Pathogenic mechanisms in ALS and FTD

Fen-Biao Gao et al

including LCD-containing proteins that mediate the formation of membraneless organelles such as SGs, suggesting a direct role in SG dynamics. Indeed, biochemical studies indicate that LCDs are direct binding targets of poly(PR) and this interaction is polymerdependent (Lin et al, 2016). Ectopic overexpression of poly(GR) or poly(PR) induces spontaneous SG assembly, a process that requires both elF2a phosphorylation and G3BP (Lee et al, 2016; Boeynaems et al, 2017). Moreover, poly(GR) and poly(PR) themselves undergo liquid–liquid phase separation in vitro and enhance the multivalent interactions of the liquid phase of SGs in living cells (Lee et al, 2016; Boeynaems et al, 2017). Since poly (PR) is not detectable in SGs (Lee et al, 2016), it will be interesting to investigate the molecular mechanism by which argininerich DPR proteins alter SG dynamics. RNA or protein products of expanded G4C2 repeats may affect the normal function of other neuronal RNP granules as well (Burguete et al, 2015). It will be important to further investigate the relationship between endogenous DPR proteins and dynamics of various RNP granules in neurons of C9ORF72 patients.

Conclusion During the last decade or so, tremendous progress has been made in understanding the common pathogenic mechanisms in both ALS and FTD. The identification of disease genes has led to breathtaking advances in knowledge about underlying molecular and cellular pathways by both translational and basic scientists in multiple related fields. As in better studied neurodegenerative diseases, it is certain that many other dysregulated common downstream pathways in the ALS-FTD spectrum disorder besides those described here will be identified in the foreseeable future. For the sake of therapy development, it is important to determine which pathway or pathways drive disease initiation and how different pathways affect each other during disease progression. Many current studies rely on overexpression of disease RNA or proteins in cellular or animal models. However, the extent to which molecular insights obtained from these studies can be translated in patient situations remains to be seen. More effort should be focused on studying the detrimental effects of disease genes expressed in their native genetic and molecular contexts. Finally, it is still not known how same mutations such as C9ORF72 repeat expansion cause distinct clinical phenotypes in different patients or why their penetrance differs in various populations. Unique genetic modifiers or environmental influences remain to be identified in years to come.

Acknowledgements We thank Li Qiu for help with EndNote. This work is supported by grants from the National Institutes of Health (R01NS057553 and R01NS079725 to F.-B.G.). F.-B.G. is also supported by the MDA Foundation, the Packard Center for ALS Research, and the Target ALS Foundation. S.A. is supported by the ALS Association, the Frick Foundation for ALS Research, and the Alzheimer’s Association (NIRG-396129).

Conflict of interest The authors declare that they have no conflict of interest.

ª 2017 The Authors

Published online: September 15, 2017

Fen-Biao Gao et al

The EMBO Journal

Pathogenic mechanisms in ALS and FTD

References

Behrends C, Sowa ME, Gygi SP, Harper JW (2010) Network organization of

Almeida S, Gascon E, Tran H, Chou HJ, Gendron TF, Degroot S, Tapper AR,

Belzil VV, Bauer PO, Prudencio M, Gendron TF, Stetler CT, Yan IK, Pregent L,

the human autophagy system. Nature 466: 68 – 76 Sellier C, Charlet-Berguerand N, Karydas A, Seeley WW, Boxer AL, Petrucelli

Daughrity L, Baker MC, Rademakers R, Boylan K, Patel TC, Dickson DW,

L, Miller BL, Gao FB (2013) Modeling key pathological features of

Petrucelli L (2013) Reduced C9orf72 gene expression in c9FTD/ALS is

frontotemporal dementia with C9ORF72 repeat expansion in iPSC-derived

caused by histone trimethylation, an epigenetic event detectable in blood.

human neurons. Acta Neuropathol 126: 385 – 399 Almeida S, Gao FB (2016) Lost & found: C9ORF72 and the autophagy pathway in ALS/FTD. EMBO J 35: 1251 – 1253 Amick J, Roczniak-Ferguson A, Ferguson SM (2016) C9orf72 binds SMCR8, localizes to lysosomes, and regulates mTORC1 signaling. Mol Biol Cell 27: 3040 – 3051 Andersson MK, Stahlberg A, Arvidsson Y, Olofsson A, Semb H, Stenman G, Nilsson O, Aman P (2008) The multifunctional FUS, EWS and TAF15 protooncoproteins show cell type-specific expression patterns and involvement in cell spreading and stress response. BMC Cell Biol 9: 37 Arai T, Hasegawa M, Akiyama H, Ikeda K, Nonaka T, Mori H, Mann D, Tsuchiya K, Yoshida M, Hashizume Y, Oda T (2006) TDP-43 is a

Acta Neuropathol 126: 895 – 905 Bentmann E, Neumann M, Tahirovic S, Rodde R, Dormann D, Haass C (2012) Requirements for stress granule recruitment of fused in sarcoma (FUS) and TAR DNA-binding protein of 43 kDa (TDP-43). J Biol Chem 287: 23079 – 23094 van Blitterswijk M, van Es MA, Hennekam EA, Dooijes D, van Rheenen W, Medic J, Bourque PR, Schelhaas HJ, van der Kooi AJ, de Visser M, de Bakker PI, Veldink JH, van den Berg LH (2012) Evidence for an oligogenic basis of amyotrophic lateral sclerosis. Hum Mol Genet 21: 3776 – 3784 van Blitterswijk M, Gendron TF, Baker MC, DeJesus-Hernandez M, Finch NA, Brown PH, Daughrity LM, Murray ME, Heckman MG, Jiang J, LagierTourenne C, Edbauer D, Cleveland DW, Josephs KA, Parisi JE, Knopman DS,

component of ubiquitin-positive tau-negative inclusions in frontotemporal

Petersen RC, Petrucelli L, Boeve BF, Graff-Radford NR et al (2015) Novel

lobar degeneration and amyotrophic lateral sclerosis. Biochem Biophys Res

clinical associations with specific C9ORF72 transcripts in patients with

Commun 351: 602 – 611 Arnold ES, Ling SC, Huelga SC, Lagier-Tourenne C, Polymenidou M, Ditsworth

repeat expansions in C9ORF72. Acta Neuropathol 130: 863 – 876 Boeynaems S, Bogaert E, Michiels E, Gijselinck I, Sieben A, Jovicic A, De Baets

D, Kordasiewicz HB, McAlonis-Downes M, Platoshyn O, Parone PA, Da Cruz

G, Scheveneels W, Steyaert J, Cuijt I, Verstrepen KJ, Callaerts P, Rousseau F,

S, Clutario KM, Swing D, Tessarollo L, Marsala M, Shaw CE, Yeo GW,

Schymkowitz J, Cruts M, Van Broeckhoven C, Van Damme P, Gitler AD,

Cleveland DW (2013) ALS-linked TDP-43 mutations produce aberrant RNA

Robberecht W, Van Den Bosch L (2016) Drosophila screen connects nuclear

splicing and adult-onset motor neuron disease without aggregation or loss of nuclear TDP-43. Proc Natl Acad Sci USA 110: E736 – E745 Arrasate M, Mitra S, Schweitzer ES, Segal MR, Finkbeiner S (2004) Inclusion

transport genes to DPR pathology in c9ALS/FTD. Sci Rep 6: 20877 Boeynaems S, Bogaert E, Kovacs D, Konijnenberg A, Timmerman E, Volkov A, Guharoy M, De Decker M, Jaspers T, Ryan VH, Janke AM, Baatsen P,

body formation reduces levels of mutant huntingtin and the risk of

Vercruysse T, Kolaitis RM, Daelemans D, Taylor JP, Kedersha N, Anderson P,

neuronal death. Nature 431: 805 – 810

Impens F, Sobott F et al (2017) Phase separation of C9orf72 dipeptide

Ash PE, Bieniek KF, Gendron TF, Caulfield T, Lin WL, Dejesus-Hernandez M, van Blitterswijk MM, Jansen-West K, Paul JW III, Rademakers R, Boylan KB,

repeats perturbs stress granule dynamics. Mol Cell 65: 1044 – 1055 Bosco DA, Lemay N, Ko HK, Zhou H, Burke C, Kwiatkowski TJ Jr, Sapp P,

Dickson DW, Petrucelli L (2013) Unconventional translation of C9ORF72

McKenna-Yasek D, Brown RH Jr, Hayward LJ (2010) Mutant FUS proteins

GGGGCC expansion generates insoluble polypeptides specific to c9FTD/ALS.

that cause amyotrophic lateral sclerosis incorporate into stress granules.

Neuron 77: 639 – 646 Atanasio A, Decman V, White D, Ramos M, Ikiz B, Lee HC, Siao CJ, Brydges S,

Hum Mol Genet 19: 4160 – 4175 Brannan KW, Jin W, Huelga SC, Banks CA, Gilmore JM, Florens L, Washburn

LaRosa E, Bai Y, Fury W, Burfeind P, Zamfirova R, Warshaw G, Orengo J,

MP, Van Nostrand EL, Pratt GA, Schwinn MK, Daniels DL, Yeo GW (2016)

Oyejide A, Fralish M, Auerbach W, Poueymirou W, Freudenberg J et al

SONAR discovers RNA-binding proteins from analysis of large-scale

(2016) C9orf72 ablation causes immune dysregulation characterized by leukocyte expansion, autoantibody production, and glomerulonephropathy in mice. Sci Rep 6: 23204 Aulas A, Stabile S, Vande Velde C (2012) Endogenous TDP-43, but not FUS, contributes to stress granule assembly via G3BP. Mol Neurodegener 7: 54 Baechtold H, Kuroda M, Sok J, Ron D, Lopez BS, Akhmedov AT (1999) Human 75-kDa DNA-pairing protein is identical to the pro-oncoprotein TLS/FUS and is able to promote D-loop formation. J Biol Chem 274: 34337 – 34342 Bannwarth S, Ait-El-Mkadem S, Chaussenot A, Genin EC, Lacas-Gervais S, Fragaki K, Berg-Alonso L, Kageyama Y, Serre V, Moore DG, Verschueren A, Rouzier C, Le Ber I, Auge G, Cochaud C, Lespinasse F, N’Guyen K, de Septenville A, Brice A, Yu-Wai-Man P et al (2014) A mitochondrial origin for frontotemporal dementia and amyotrophic lateral sclerosis through CHCHD10 involvement. Brain 137: 2329 – 2345 Beck M, Hurt E (2017) The nuclear pore complex: understanding its function through structural insight. Nat Rev Mol Cell Biol 18: 73 – 89 Becker LA, Huang B, Bieri G, Ma R, Knowles DA, Jafar-Nejad P, Messing J, Kim HJ, Soriano A, Auburger G, Pulst SM, Taylor JP, Rigo F, Gitler AD (2017)

protein-protein interactomes. Mol Cell 64: 282 – 293 Brenner D, Muller K, Wieland T, Weydt P, Bohm S, Lule D, Hubers A, Neuwirth C, Weber M, Borck G, Wahlqvist M, Danzer KM, Volk AE, Meitinger T, Strom TM, Otto M, Kassubek J, Ludolph AC, Andersen PM, Weishaupt JH (2016) NEK1 mutations in familial amyotrophic lateral sclerosis. Brain 139: e28 Britton S, Dernoncourt E, Delteil C, Froment C, Schiltz O, Salles B, Frit P, Calsou P (2014) DNA damage triggers SAF-A and RNA biogenesis factors exclusion from chromatin coupled to R-loops removal. Nucleic Acids Res 42: 9047 – 9062 Brown RH Jr (1997) Amyotrophic lateral sclerosis. Insights from genetics. Arch Neurol 54: 1246 – 1250 Buchan JR, Kolaitis RM, Taylor JP, Parker R (2013) Eukaryotic stress granules are cleared by autophagy and Cdc48/VCP function. Cell 153: 1461 – 1474 Buratti E, Dork T, Zuccato E, Pagani F, Romano M, Baralle FE (2001) Nuclear factor TDP-43 and SR proteins promote in vitro and in vivo CFTR exon 9 skipping. EMBO J 20: 1774 – 1784 Burberry A, Suzuki N, Wang JY, Moccia R, Mordes DA, Stewart MH, Suzuki-

Therapeutic reduction of ataxin-2 extends lifespan and reduces pathology

Uematsu S, Ghosh S, Singh A, Merkle FT, Koszka K, Li QZ, Zon L, Rossi DJ,

in TDP-43 mice. Nature 544: 367 – 371

Trowbridge JJ, Notarangelo LD, Eggan K (2016) Loss-of-function mutations

ª 2017 The Authors

The EMBO Journal

11

Published online: September 15, 2017

The EMBO Journal

in the C9ORF72 mouse ortholog cause fatal autoimmune disease. Sci Transl Med 8: 347ra93 Burguete AS, Almeida S, Gao FB, Kalb R, Akins MR, Bonini NM (2015) GGGGCC microsatellite RNA is neuritically localized, induces branching defects, and perturbs transport granule function. Elife 4: e08881 Burke KA, Janke AM, Rhine CL, Fawzi NL (2015) Residue-by-residue view of

Fen-Biao Gao et al

Cox LE, Ferraiuolo L, Goodall EF, Heath PR, Higginbottom A, Mortiboys H, Hollinger HC, Hartley JA, Brockington A, Burness CE, Morrison KE, Wharton SB, Grierson AJ, Ince PG, Kirby J, Shaw PJ (2010) Mutations in CHMP2B in lower motor neuron predominant amyotrophic lateral sclerosis (ALS). PLoS ONE 5: e9872 Daigle JG, Lanson NA Jr, Smith RB, Casci I, Maltare A, Monaghan J, Nichols

in vitro FUS granules that bind the C-terminal domain of RNA polymerase

CD, Kryndushkin D, Shewmaker F, Pandey UB (2013) RNA-binding ability

II. Mol Cell 60: 231 – 241

of FUS regulates neurodegeneration, cytoplasmic mislocalization and

Chabot B, Shkreta L (2016) Defective control of pre-messenger RNA splicing in human disease. J Cell Biol 212: 13 – 27 Chang L, Monteiro MJ (2015) Defective proteasome delivery of polyubiquitinated proteins by ubiquilin-2 proteins containing ALS mutations. PLoS ONE 10: e0130162 Chew J, Gendron TF, Prudencio M, Sasaguri H, Zhang YJ, Castanedes-Casey M,

incorporation into stress granules associated with FUS carrying ALS-linked mutations. Hum Mol Genet 22: 1193 – 1205 D’Angelo MA, Raices M, Panowski SH, Hetzer MW (2009) Age-dependent deterioration of nuclear pore complexes causes a loss of nuclear integrity in postmitotic cells. Cell 136: 284 – 295 DeJesus-Hernandez M, Mackenzie IR, Boeve BF, Boxer AL, Baker M, Rutherford

Lee CW, Jansen-West K, Kurti A, Murray ME, Bieniek KF, Bauer PO,

NJ, Nicholson AM, Finch NA, Flynn H, Adamson J, Kouri N, Wojtas A,

Whitelaw EC, Rousseau L, Stankowski JN, Stetler C, Daughrity LM,

Sengdy P, Hsiung GY, Karydas A, Seeley WW, Josephs KA, Coppola G,

Perkerson EA, Desaro P, Johnston A et al (2015) Neurodegeneration.

Geschwind DH, Wszolek ZK et al (2011) Expanded GGGGCC

C9ORF72 repeat expansions in mice cause TDP-43 pathology, neuronal

hexanucleotide repeat in noncoding region of C9ORF72 causes

loss, and behavioral deficits. Science 348: 1151 – 1154 Ching JK, Elizabeth SV, Ju JS, Lusk C, Pittman SK, Weihl CC (2013) mTOR

chromosome 9p-linked FTD and ALS. Neuron 72: 245 – 256 DeJesus-Hernandez M, Finch NA, Wang X, Gendron TF, Bieniek KF, Heckman

dysfunction contributes to vacuolar pathology and weakness in valosin-

MG, Vasilevich A, Murray ME, Rousseau L, Weesner R, Lucido A, Parsons M,

containing protein associated inclusion body myopathy. Hum Mol Genet

Chew J, Josephs KA, Parisi JE, Knopman DS, Petersen RC, Boeve BF, Graff-

22: 1167 – 1179 Cirulli ET, Lasseigne BN, Petrovski S, Sapp PC, Dion PA, Leblond CS, Couthouis J, Lu YF, Wang Q, Krueger BJ, Ren Z, Keebler J, Han Y, Levy SE, Boone BE,

Radford NR, de Boer J et al (2017) In-depth clinico-pathological examination of RNA foci in a large cohort of C9ORF72 expansion carriers. Acta Neuropathol 134: 255 – 269

Wimbish JR, Waite LL, Jones AL, Carulli JP, Day-Williams AG et al (2015)

Deng HX, Chen W, Hong ST, Boycott KM, Gorrie GH, Siddique N, Yang Y,

Exome sequencing in amyotrophic lateral sclerosis identifies risk genes

Fecto F, Shi Y, Zhai H, Jiang H, Hirano M, Rampersaud E, Jansen GH,

and pathways. Science 347: 1436 – 1441

Donkervoort S, Bigio EH, Brooks BR, Ajroud K, Sufit RL, Haines JL et al

Clayton EL, Mizielinska S, Edgar JR, Nielsen TT, Marshall S, Norona FE, Robbins M, Damirji H, Holm IE, Johannsen P, Nielsen JE, Asante EA, Collinge J, FReJA Consortium, Isaacs AM (2015) Frontotemporal dementia

(2011) Mutations in UBQLN2 cause dominant X-linked juvenile and adultonset ALS and ALS/dementia. Nature 477: 211 – 215 Deng Q, Holler CJ, Taylor G, Hudson KF, Watkins W, Gearing M, Ito D, Murray

caused by CHMP2B mutation is characterised by neuronal lysosomal

ME, Dickson DW, Seyfried NT, Kukar T (2014) FUS is phosphorylated by

storage pathology. Acta Neuropathol 130: 511 – 523

DNA-PK and accumulates in the cytoplasm after DNA damage. J Neurosci

Clayton EL, Mancuso R, Nielsen TT, Mizielinska S, Holmes H, Powell N, Norona F, Overgaard Larsen J, Milioto C, Wilson KM, Lythgoe MF, Ourselin

34: 7802 – 7813 Dewey CM, Cenik B, Sephton CF, Dries DR, Mayer P III, Good SK, Johnson BA,

S, Nielsen JE, Johannsen P, Holm I, Collinge J, Frej A, Oliver PL, Gomez-

Herz J, Yu G (2011) TDP-43 is directed to stress granules by sorbitol, a novel

Nicola D, Isaacs AM (2017) Early microgliosis precedes neuronal loss and

physiological osmotic and oxidative stressor. Mol Cell Biol 31: 1098 – 1108

behavioural impairment in mice with a frontotemporal dementia-causing CHMP2B mutation. Hum Mol Genet 26: 873 – 887 Colombrita C, Onesto E, Megiorni F, Pizzuti A, Baralle FE, Buratti E, Silani V,

Donnelly CJ, Zhang PW, Pham JT, Haeusler AR, Mistry NA, Vidensky S, Daley EL, Poth EM, Hoover B, Fines DM, Maragakis N, Tienari PJ, Petrucelli L, Traynor BJ, Wang J, Rigo F, Bennett CF, Blackshaw S, Sattler R, Rothstein

Ratti A (2012) TDP-43 and FUS RNA-binding proteins bind distinct sets of

JD (2013) RNA toxicity from the ALS/FTD C9ORF72 expansion is mitigated

cytoplasmic messenger RNAs and differently regulate their post-

by antisense intervention. Neuron 80: 415 – 428

transcriptional fate in motoneuron-like cells. J Biol Chem 287: 15635 – 15647 Conlon EG, Lu L, Sharma A, Yamazaki T, Tang T, Shneider NA, Manley JL (2016) The C9ORF72 GGGGCC expansion forms RNA G-quadruplex inclusions and sequesters hnRNP H to disrupt splicing in ALS brains. Elife 5: e17820 Cooper-Knock J, Walsh MJ, Higginbottom A, Robin Highley J, Dickman MJ, Edbauer D, Ince PG, Wharton SB, Wilson SA, Kirby J, Hautbergue GM,

Dormann D, Rodde R, Edbauer D, Bentmann E, Fischer I, Hruscha A, Than ME, Mackenzie IR, Capell A, Schmid B, Neumann M, Haass C (2010) ALSassociated fused in sarcoma (FUS) mutations disrupt Transportinmediated nuclear import. EMBO J 29: 2841 – 2857 Fan Z, Chen X, Chen R (2014) Transcriptome-wide analysis of TDP-43 binding small RNAs identifies miR-NID1 (miR-8485), a novel miRNA that represses NRXN1 expression. Genomics 103: 76 – 82 Farg MA, Sundaramoorthy V, Sultana JM, Yang S, Atkinson RA, Levina V,

Shaw PJ (2014) Sequestration of multiple RNA recognition motif-

Halloran MA, Gleeson PA, Blair IP, Soo KY, King AE, Atkin JD (2014)

containing proteins by C9orf72 repeat expansions. Brain 137:

C9ORF72, implicated in amytrophic lateral sclerosis and frontotemporal

2040 – 2051 Cooper-Knock J, Bury JJ, Heath PR, Wyles M, Higginbottom A, Gelsthorpe C, Highley JR, Hautbergue G, Rattray M, Kirby J, Shaw PJ (2015) C9ORF72 GGGGCC expanded repeats produce splicing dysregulation which correlates with disease severity in amyotrophic lateral sclerosis. PLoS ONE 10: e0127376

12

Pathogenic mechanisms in ALS and FTD

The EMBO Journal

dementia, regulates endosomal trafficking. Hum Mol Genet 23: 3579 – 3595 Farg MA, Konopka A, Ying Soo K, Ito D, Atkin JD (2017) The DNA damage response (DDR) is induced by the C9orf72 repeat expansion in amyotrophic lateral sclerosis. Hum Mol Genet 26: 2882 – 2896 Fecto F, Yan J, Vemula SP, Liu E, Yang Y, Chen W, Zheng JG, Shi Y, Siddique N, Arrat H, Donkervoort S, Ajroud-Driss S, Sufit RL, Heller SL, Deng HX,

ª 2017 The Authors

Published online: September 15, 2017

Fen-Biao Gao et al

The EMBO Journal

Pathogenic mechanisms in ALS and FTD

Siddique T (2011) SQSTM1 mutations in familial and sporadic amyotrophic lateral sclerosis. Arch Neurol 68: 1440 – 1446 Filimonenko M, Stuffers S, Raiborg C, Yamamoto A, Malerod L, Fisher EM, Isaacs A, Brech A, Stenmark H, Simonsen A (2007) Functional multivesicular bodies are required for autophagic clearance of protein aggregates associated with neurodegenerative disease. J Cell Biol 179: 485 – 500 Freibaum BD, Chitta RK, High AA, Taylor JP (2010) Global analysis of TDP-43

Goode A, Rea S, Sultana M, Shaw B, Searle MS, Layfield R (2016b) ALS-FTLD associated mutations of SQSTM1 impact on Keap1-Nrf2 signalling. Mol Cell Neurosci 76: 52 – 58 Gorlich D, Kutay U (1999) Transport between the cell nucleus and the cytoplasm. Annu Rev Cell Dev Biol 15: 607 – 660 Grima JC, Daigle JG, Arbez N, Cunningham KC, Zhang K, Ochaba J, Geater C, Morozko E, Stocksdale J, Glatzer JC, Pham JT, Ahmed I, Peng Q, Wadhwa H, Pletnikova O, Troncoso JC, Duan W, Snyder SH, Ranum LP, Thompson

interacting proteins reveals strong association with RNA splicing and

LM et al (2017) Mutant huntingtin disrupts the nuclear pore complex.

translation machinery. J Proteome Res 9: 1104 – 1120

Neuron 94: 93 – 107

Freibaum BD, Lu Y, Lopez-Gonzalez R, Kim NC, Almeida S, Lee KH, Badders N,

Haeusler AR, Donnelly CJ, Periz G, Simko EA, Shaw PG, Kim MS, Maragakis NJ,

Valentine M, Miller BL, Wong PC, Petrucelli L, Kim HJ, Gao FB, Taylor JP

Troncoso JC, Pandey A, Sattler R, Rothstein JD, Wang J (2014) C9orf72

(2015) GGGGCC repeat expansion in C9orf72 compromises

nucleotide repeat structures initiate molecular cascades of disease. Nature

nucleocytoplasmic transport. Nature 525: 129 – 133 Freischmidt A, Wieland T, Richter B, Ruf W, Schaeffer V, Muller K, Marroquin

507: 195 – 200 Heo JM, Ordureau A, Paulo JA, Rinehart J, Harper JW (2015) The PINK1-

N, Nordin F, Hubers A, Weydt P, Pinto S, Press R, Millecamps S, Molko N,

PARKIN mitochondrial ubiquitylation pathway drives a program of OPTN/

Bernard E, Desnuelle C, Soriani MH, Dorst J, Graf E, Nordstrom U et al

NDP52 recruitment and TBK1 activation to promote mitophagy. Mol Cell

(2015) Haploinsufficiency of TBK1 causes familial ALS and fronto-temporal dementia. Nat Neurosci 18: 631 – 636 Gal J, Zhang J, Kwinter DM, Zhai J, Jia H, Jia J, Zhu H (2011) Nuclear localization sequence of FUS and induction of stress granules by ALS mutants. Neurobiol Aging 32: 2323.e27 – 2323.e40 Gao FB, Richter JD (2017) Microsatellite expansion diseases: repeat toxicity found in translation. Neuron 93: 249 – 251 Gascon E, Lynch K, Ruan H, Almeida S, Verheyden JM, Seeley WW, Dickson DW, Petrucelli L, Sun D, Jiao J, Zhou H, Jakovcevski M, Akbarian S, Yao WD, Gao FB (2014) Alterations in microRNA-124 and AMPA receptors contribute to social behavioral deficits in frontotemporal dementia. Nat Med 20: 1444 – 1451 Gasset-Rosa F, Chillon-Marinas C, Goginashvili A, Atwal RS, Artates JW, Tabet R, Wheeler VC, Bang AG, Cleveland DW, Lagier-Tourenne C (2017) Polyglutamine-expanded huntingtin exacerbates age-related disruption of nuclear integrity and nucleocytoplasmic transport. Neuron 94: 48 – 57 Gendron TF, Bieniek KF, Zhang YJ, Jansen-West K, Ash PE, Caulfield T,

60: 7 – 20 Hicks GG, Singh N, Nashabi A, Mai S, Bozek G, Klewes L, Arapovic D, White EK, Koury MJ, Oltz EM, Van Kaer L, Ruley HE (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 Higelin J, Demestre M, Putz S, Delling JP, Jacob C, Lutz AK, Bausinger J, Huber AK, Klingenstein M, Barbi G, Speit G, Huebers A, Weishaupt JH, Hermann A, Liebau S, Ludolph AC, Boeckers TM (2016) FUS mislocalization and vulnerability to DNA damage in ALS patients derived hiPSCs and aging motoneurons. Front Cell Neurosci 10: 290 Hill SJ, Mordes DA, Cameron LA, Neuberg DS, Landini S, Eggan K, Livingston DM (2016) Two familial ALS proteins function in prevention/repair of transcription-associated DNA damage. Proc Natl Acad Sci USA 113: E7701 – E7709 Hjerpe R, Bett JS, Keuss MJ, Solovyova A, McWilliams TG, Johnson C, Sahu I, Varghese J, Wood N, Wightman M, Osborne G, Bates GP, Glickman MH, Trost M, Knebel A, Marchesi F, Kurz T (2016) UBQLN2 mediates

Daughrity L, Dunmore JH, Castanedes-Casey M, Chew J, Cosio DM, van

autophagy-independent protein aggregate clearance by the proteasome.

Blitterswijk M, Lee WC, Rademakers R, Boylan KB, Dickson DW, Petrucelli L

Cell 166: 935 – 949

(2013) Antisense transcripts of the expanded C9ORF72 hexanucleotide repeat form nuclear RNA foci and undergo repeat-associated non-ATG translation in c9FTD/ALS. Acta Neuropathol 126: 829 – 844 Gendron TF, Petrucelli L (2017) Disease mechanisms of C9ORF72 repeat expansions. Cold Spring Harb Perspect Med https://doi.org/10.1101/cshpe rspect.a024224 Ghazi-Noori S, Froud KE, Mizielinska S, Powell C, Smidak M, Fernandez de Marco M, O’Malley C, Farmer M, Parkinson N, Fisher EM, Asante EA, Brandner S, Collinge J, Isaacs AM (2012) Progressive neuronal inclusion formation and axonal degeneration in CHMP2B mutant transgenic mice. Brain 135: 819 – 832 Gijselinck I, Van Langenhove T, van der Zee J, Sleegers K, Philtjens S,

Hong S, Dissing-Olesen L, Stevens B (2016) New insights on the role of microglia in synaptic pruning in health and disease. Curr Opin Neurobiol 36: 128 – 134 Horoupian DS, Thal L, Katzman R, Terry RD, Davies P, Hirano A, DeTeresa R, Fuld PA, Petito C, Blass J, Ellis JM (1984) Dementia and motor neuron disease: morphometric, biochemical, and Golgi studies. Ann Neurol 16: 305 – 313 Hudson AJ (1981) Amyotrophic lateral sclerosis and its association with dementia, parkinsonism and other neurological disorders: a review. Brain 104: 217 – 247 Jain A, Vale RD (2017) RNA phase transitions in repeat expansion disorders. Nature 546: 243 – 247 Japtok J, Lojewski X, Naumann M, Klingenstein M, Reinhardt P, Sterneckert J,

Kleinberger G, Janssens J, Bettens K, Van Cauwenberghe C, Pereson S,

Putz S, Demestre M, Boeckers TM, Ludolph AC, Liebau S, Storch A,

Engelborghs S, Sieben A, De Jonghe P, Vandenberghe R, Santens P, De

Hermann A (2015) Stepwise acquirement of hallmark neuropathology in

Bleecker J, Maes G, Baumer V, Dillen L, Joris G et al (2012) A C9orf72

FUS-ALS iPSC models depends on mutation type and neuronal aging.

promoter repeat expansion in a Flanders-Belgian cohort with disorders of the frontotemporal lobar degeneration-amyotrophic lateral sclerosis spectrum: a gene identification study. Lancet Neurol 11: 54 – 65 Goode A, Butler K, Long J, Cavey J, Scott D, Shaw B, Sollenberger J, Gell C, Johansen T, Oldham NJ, Searle MS, Layfield R (2016a) Defective recognition

Neurobiol Dis 82: 420 – 429 Jeong YH, Ling JP, Lin SZ, Donde AN, Braunstein KE, Majounie E, Traynor BJ, LaClair KD, Lloyd TE, Wong PC (2017) Tdp-43 cryptic exons are highly variable between cell types. Mol Neurodegener 12: 13 Jiang J, Zhu Q, Gendron TF, Saberi S, McAlonis-Downes M, Seelman A,

of LC3B by mutant SQSTM1/p62 implicates impairment of autophagy as a

Stauffer JE, Jafar-Nejad P, Drenner K, Schulte D, Chun S, Sun S, Ling SC,

pathogenic mechanism in ALS-FTLD. Autophagy 12: 1094 – 1104

Myers B, Engelhardt J, Katz M, Baughn M, Platoshyn O, Marsala M, Watt A

ª 2017 The Authors

The EMBO Journal

13

Published online: September 15, 2017

The EMBO Journal

et al (2016) Gain of toxicity from ALS/FTD-linked repeat expansions in C9ORF72 is alleviated by antisense oligonucleotides targeting GGGGCCcontaining RNAs. Neuron 90: 535 – 550 Johnson JO, Mandrioli J, Benatar M, Abramzon Y, Van Deerlin VM, Trojanowski JQ, Gibbs JR, Brunetti M, Gronka S, Wuu J, Ding J, McCluskey L, MartinezLage M, Falcone D, Hernandez DG, Arepalli S, Chong S, Schymick JC, Rothstein J, Landi F et al (2010) Exome sequencing reveals VCP mutations as a cause of familial ALS. Neuron 68: 857 – 864 Jovicic A, Mertens J, Boeynaems S, Bogaert E, Chai N, Yamada SB, Paul JW III, Sun S, Herdy JR, Bieri G, Kramer NJ, Gage FH, Van Den Bosch L, Robberecht W, Gitler AD (2015) Modifiers of C9orf72 dipeptide repeat toxicity connect nucleocytoplasmic transport defects to FTD/ALS. Nat Neurosci 18: 1226 – 1229 Ju JS, Miller SE, Hanson PI, Weihl CC (2008) Impaired protein aggregate handling and clearance underlie the pathogenesis of p97/VCP-associated disease. J Biol Chem 283: 30289 – 30299 Ju JS, Fuentealba RA, Miller SE, Jackson E, Piwnica-Worms D, Baloh RH, Weihl CC (2009) Valosin-containing protein (VCP) is required for autophagy and is disrupted in VCP disease. J Cell Biol 187: 875 – 888 Jung J, Nayak A, Schaeffer V, Starzetz T, Kirsch AK, Muller S, Dikic I,

Fen-Biao Gao et al

Kim HJ, Raphael AR, LaDow ES, McGurk L, Weber RA, Trojanowski JQ, Lee VM, Finkbeiner S, Gitler AD, Bonini NM (2014) Therapeutic modulation of eIF2alpha phosphorylation rescues TDP-43 toxicity in amyotrophic lateral sclerosis disease models. Nat Genet 46: 152 – 160 King OD, Gitler AD, Shorter J (2012) The tip of the iceberg: RNA-binding proteins with prion-like domains in neurodegenerative disease. Brain Res 1462: 61 – 80 Kinoshita Y, Ito H, Hirano A, Fujita K, Wate R, Nakamura M, Kaneko S, Nakano S, Kusaka H (2009) Nuclear contour irregularity and abnormal transporter protein distribution in anterior horn cells in amyotrophic lateral sclerosis. J Neuropathol Exp Neurol 68: 1184 – 1192 Koppers M, Blokhuis AM, Westeneng HJ, Terpstra ML, Zundel CA, Vieira de Sa R, Schellevis RD, Waite AJ, Blake DJ, Veldink JH, van den Berg LH, Pasterkamp RJ (2015) C9orf72 ablation in mice does not cause motor neuron degeneration or motor deficits. Ann Neurol 78: 426 – 438 Korac J, Schaeffer V, Kovacevic I, Clement AM, Jungblut B, Behl C, Terzic J, Dikic I (2013) Ubiquitin-independent function of optineurin in autophagic clearance of protein aggregates. J Cell Sci 126: 580 – 592 Krug L, Chatterjee N, Borges-Monroy R, Hearn S, Liao WW, Morrill K, Prazak L, Rozhkov N, Theodorou D, Hammell M, Dubnau J (2017) Retrotransposon

Mittelbronn M, Behrends C (2017) Multiplex image-based autophagy RNAi

activation contributes to neurodegeneration in a Drosophila TDP-43 model

screening identifies SMCR8 as ULK1 kinase activity and gene expression

of ALS. PLoS Genet 13: e1006635

regulator. Elife 6: e23063 Kabashi E, Valdmanis PN, Dion P, Spiegelman D, McConkey BJ, Vande Velde C, Bouchard JP, Lacomblez L, Pochigaeva K, Salachas F, Pradat PF, Camu W, Meininger V, Dupre N, Rouleau GA (2008) TARDBP mutations in

Kuroda M, Sok J, Webb L, Baechtold H, Urano F, Yin Y, Chung P, de Rooij DG, Akhmedov A, Ashley T, Ron D (2000) Male sterility and enhanced radiation sensitivity in TLS–/– mice. EMBO J 19: 453 – 462 Kwiatkowski TJ Jr, Bosco DA, Leclerc AL, Tamrazian E, Vanderburg CR, Russ C,

individuals with sporadic and familial amyotrophic lateral sclerosis. Nat

Davis A, Gilchrist J, Kasarskis EJ, Munsat T, Valdmanis P, Rouleau GA,

Genet 40: 572 – 574

Hosler BA, Cortelli P, de Jong PJ, Yoshinaga Y, Haines JL, Pericak-Vance MA,

Kaneb HM, Folkmann AW, Belzil VV, Jao LE, Leblond CS, Girard SL, Daoud H, Noreau A, Rochefort D, Hince P, Szuto A, Levert A, Vidal S, Andre-Guimont C, Camu W, Bouchard JP, Dupre N, Rouleau GA, Wente SR, Dion PA (2015) Deleterious mutations in the essential mRNA metabolism factor, hGle1, in amyotrophic lateral sclerosis. Hum Mol Genet 24: 1363 – 1373

Yan J, Ticozzi N et al (2009) Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science 323: 1205 – 1208 Lagier-Tourenne C, Polymenidou M, Hutt KR, Vu AQ, Baughn M, Huelga SC, Clutario KM, Ling SC, Liang TY, Mazur C, Wancewicz E, Kim AS,

Kanekura K, Yagi T, Cammack AJ, Mahadevan J, Kuroda M, Harms MB, Miller

Watt A, Freier S, Hicks GG, Donohue JP, Shiue L, Bennett CF, Ravits J,

TM, Urano F (2016) Poly-dipeptides encoded by the C9ORF72 repeats

Cleveland DW et al (2012) Divergent roles of ALS-linked proteins FUS/

block global protein translation. Hum Mol Genet 25: 1803 – 1813 Kao AW, McKay A, Singh PP, Brunet A, Huang EJ (2017) Progranulin, lysosomal regulation and neurodegenerative disease. Nat Rev Neurosci 18: 325 – 333 Kenna KP, van Doormaal PT, Dekker AM, Ticozzi N, Kenna BJ, Diekstra FP, van Rheenen W, van Eijk KR, Jones AR, Keagle P, Shatunov A, Sproviero W,

TLS and TDP-43 intersect in processing long pre-mRNAs. Nat Neurosci 15: 1488 – 1497 Lagier-Tourenne C, Baughn M, Rigo F, Sun S, Liu P, Li HR, Jiang J, Watt AT, Chun S, Katz M, Qiu J, Sun Y, Ling SC, Zhu Q, Polymenidou M, Drenner K, Artates JW, McAlonis-Downes M, Markmiller S, Hutt KR et al (2013) Targeted degradation of sense and antisense C9orf72 RNA foci as therapy

Smith BN, van Es MA, Topp SD, Kenna A, Miller JW, Fallini C, Tiloca C,

for ALS and frontotemporal degeneration. Proc Natl Acad Sci USA 110:

McLaughlin RL et al (2016) NEK1 variants confer susceptibility to

E4530 – E4539

amyotrophic lateral sclerosis. Nat Genet 48: 1037 – 1042 Khosravi B, Hartmann H, May S, Mohl C, Ederle H, Michaelsen M, Schludi MH, Dormann D, Edbauer D (2017) Cytoplasmic poly-GA aggregates impair nuclear import of TDP-43 in C9orf72 ALS/FTLD. Hum Mol Genet 26: 790 – 800 Kim HJ, Kim NC, Wang YD, Scarborough EA, Moore J, Diaz Z, MacLea KS, Freibaum B, Li S, Molliex A, Kanagaraj AP, Carter R, Boylan KB, Wojtas AM, Rademakers R, Pinkus JL, Greenberg SA, Trojanowski JQ, Traynor BJ, Smith BN et al (2013a) Mutations in prion-like domains in hnRNPA2B1 and hnRNPA1 cause multisystem proteinopathy and ALS. Nature 495: 467 – 473 Kim NC, Tresse E, Kolaitis RM, Molliex A, Thomas RE, Alami NH, Wang B, Joshi A, Smith RB, Ritson GP, Winborn BJ, Moore J, Lee JY, Yao TP, Pallanck

14

Pathogenic mechanisms in ALS and FTD

Lattante S, de Calbiac H, Le Ber I, Brice A, Ciura S, Kabashi E (2015) Sqstm1 knock-down causes a locomotor phenotype ameliorated by rapamycin in a zebrafish model of ALS/FTLD. Hum Mol Genet 24: 1682 – 1690 Le NT, Chang L, Kovlyagina I, Georgiou P, Safren N, Braunstein KE, Kvarta MD, Van Dyke AM, LeGates TA, Philips T, Morrison BM, Thompson SM, Puche AC, Gould TD, Rothstein JD, Wong PC, Monteiro MJ (2016) Motor neuron disease, TDP-43 pathology, and memory deficits in mice expressing ALSFTD-linked UBQLN2 mutations. Proc Natl Acad Sci USA 113: E7580 – E7589 Lee JA, Beigneux A, Ahmad ST, Young SG, Gao FB (2007) ESCRT-III dysfunction causes autophagosome accumulation and neurodegeneration. Curr Biol 17: 1561 – 1567 Lee YB, Chen HJ, Peres JN, Gomez-Deza J, Attig J, Stalekar M, Troakes C,

L, Kundu M, Taylor JP (2013b) VCP is essential for mitochondrial quality

Nishimura AL, Scotter EL, Vance C, Adachi Y, Sardone V, Miller JW, Smith

control by PINK1/Parkin and this function is impaired by VCP mutations.

BN, Gallo JM, Ule J, Hirth F, Rogelj B, Houart C, Shaw CE (2013)

Neuron 78: 65 – 80

Hexanucleotide repeats in ALS/FTD form length-dependent RNA foci,

The EMBO Journal

ª 2017 The Authors

Published online: September 15, 2017

Fen-Biao Gao et al

The EMBO Journal

Pathogenic mechanisms in ALS and FTD

sequester RNA binding proteins, and are neurotoxic. Cell Rep 5: 1178 – 1186

Lui H, Zhang J, Makinson SR, Cahill MK, Kelley KW, Huang HY, Shang Y, Oldham MC, Martens LH, Gao F, Coppola G, Sloan SA, Hsieh CL, Kim CC,

Lee KH, Zhang P, Kim HJ, Mitrea DM, Sarkar M, Freibaum BD, Cika J, Coughlin

Bigio EH, Weintraub S, Mesulam MM, Rademakers R, Mackenzie IR, Seeley

M, Messing J, Molliex A, Maxwell BA, Kim NC, Temirov J, Moore J, Kolaitis

WW et al (2016) Progranulin deficiency promotes circuit-specific synaptic

RM, Shaw TI, Bai B, Peng J, Kriwacki RW, Taylor JP (2016) C9orf72

pruning by microglia via complement activation. Cell 165: 921 – 935

dipeptide repeats impair the assembly, dynamics, and function of

Lukavsky PJ, Daujotyte D, Tollervey JR, Ule J, Stuani C, Buratti E, Baralle FE,

membrane-less organelles. Cell 167: 774 – 788.e17 Lenzi J, De Santis R, de Turris V, Morlando M, Laneve P, Calvo A, Caliendo V, Chio A, Rosa A, Bozzoni I (2015) ALS mutant FUS proteins are recruited into stress granules in induced pluripotent stem cell-derived motoneurons. Dis Model Mech 8: 755 – 766 Levine TP, Daniels RD, Gatta AT, Wong LH, Hayes MJ (2013) The product of

Damberger FF, Allain FH (2013) Molecular basis of UG-rich RNA recognition by the human splicing factor TDP-43. Nat Struct Mol Biol 20: 1443 – 1449 Mackenzie IR, Neumann M, Bigio EH, Cairns NJ, Alafuzoff I, Kril J, Kovacs GG, Ghetti B, Halliday G, Holm IE, Ince PG, Kamphorst W, Revesz T, Rozemuller AJ, Kumar-Singh S, Akiyama H, Baborie A, Spina S, Dickson DW,

C9orf72, a gene strongly implicated in neurodegeneration, is structurally

Trojanowski JQ et al (2010) Nomenclature and nosology for

related to DENN Rab-GEFs. Bioinformatics 29: 499 – 503

neuropathologic subtypes of frontotemporal lobar degeneration: an

Li YR, King OD, Shorter J, Gitler AD (2013a) Stress granules as crucibles of ALS pathogenesis. J Cell Biol 201: 361 – 372 Li Z, Lu Y, Xu XL, Gao FB (2013b) The FTD/ALS-associated RNA-binding protein TDP-43 regulates the robustness of neuronal specification through microRNA-9a in Drosophila. Hum Mol Genet 22: 218 – 225 Li F, Xie X, Wang Y, Liu J, Cheng X, Guo Y, Gong Y, Hu S, Pan L (2016)

update. Acta Neuropathol 119: 1 – 4 Mackenzie IR, Arzberger T, Kremmer E, Troost D, Lorenzl S, Mori K, Weng SM, Haass C, Kretzschmar HA, Edbauer D, Neumann M (2013) Dipeptide repeat protein pathology in C9ORF72 mutation cases: clinico-pathological correlations. Acta Neuropathol 126: 859 – 879 Mackenzie IR, Frick P, Grasser FA, Gendron TF, Petrucelli L, Cashman NR,

Structural insights into the interaction and disease mechanism of

Edbauer D, Kremmer E, Prudlo J, Troost D, Neumann M (2015)

neurodegenerative disease-associated optineurin and TBK1 proteins. Nat

Quantitative analysis and clinico-pathological correlations of different

Commun 7: 12708

dipeptide repeat protein pathologies in C9ORF72 mutation carriers. Acta

Lim SM, Choi WJ, Oh KW, Xue Y, Choi JY, Kim SH, Nahm M, Kim YE, Lee J,

Neuropathol 130: 845 – 861

Noh MY, Lee S, Hwang S, Ki CS, Fu XD, Kim SH (2016) Directly converted

Mackenzie IR, Nicholson AM, Sarkar M, Messing J, Purice MD, Pottier C, Annu

patient-specific induced neurons mirror the neuropathology of FUS with

K, Baker M, Perkerson RB, Kurti A, Matchett BJ, Mittag T, Temirov J, Hsiung

disrupted nuclear localization in amyotrophic lateral sclerosis. Mol

GR, Krieger C, Murray ME, Kato M, Fryer JD, Petrucelli L, Zinman L et al

Neurodegener 11: 8

(2017) TIA1 mutations in amyotrophic lateral sclerosis and frontotemporal

Lin Y, Protter DS, Rosen MK, Parker R (2015) Formation and maturation of phase-separated liquid droplets by RNA-binding proteins. Mol Cell 60: 208 – 219 Lin Y, Mori E, Kato M, Xiang S, Wu L, Kwon I, McKnight SL (2016) Toxic PR poly-dipeptides encoded by the C9orf72 repeat expansion target LC domain polymers. Cell 167: 789 – 802 Ling SC, Polymenidou M, Cleveland DW (2013) Converging mechanisms in ALS and FTD: disrupted RNA and protein homeostasis. Neuron 79: 416 – 438

dementia promote phase separation and alter stress granule dynamics. Neuron 95: 808 – 816 Madabhushi R, Pan L, Tsai LH (2014) DNA damage and its links to neurodegeneration. Neuron 83: 266 – 282 Madabhushi R, Gao F, Pfenning AR, Pan L, Yamakawa S, Seo J, Rueda R, Phan TX, Yamakawa H, Pao PC, Stott RT, Gjoneska E, Nott A, Cho S, Kellis M, Tsai LH (2015) Activity-induced DNA breaks govern the expression of neuronal early-response genes. Cell 161: 1592 – 1605 Martinez FJ, Pratt GA, Van Nostrand EL, Batra R, Huelga SC, Kapeli K, Freese

Ling JP, Pletnikova O, Troncoso JC, Wong PC (2015) TDP-43 repression of

P, Chun SJ, Ling K, Gelboin-Burkhart C, Fijany L, Wang HC, Nussbacher JK,

nonconserved cryptic exons is compromised in ALS-FTD. Science 349:

Broski SM, Kim HJ, Lardelli R, Sundararaman B, Donohue JP, Javaherian A,

650 – 655 Liu Y, Pattamatta A, Zu T, Reid T, Bardhi O, Borchelt DR, Yachnis AT, Ranum LP (2016) C9orf72 BAC mouse model with motor deficits and neurodegenerative features of ALS/FTD. Neuron 90: 521 – 934 Liu-Yesucevitz L, Bilgutay A, Zhang YJ, Vanderweyde T, Citro A, Mehta T,

Lykke-Andersen J et al (2016) Protein-RNA networks regulated by normal and ALS-associated mutant hnRNPA2B1 in the nervous system. Neuron 92: 780 – 795 Maruyama H, Morino H, Ito H, Izumi Y, Kato H, Watanabe Y, Kinoshita Y, Kamada M, Nodera H, Suzuki H, Komure O, Matsuura S, Kobatake K,

Zaarur N, McKee A, Bowser R, Sherman M, Petrucelli L, Wolozin B (2010)

Morimoto N, Abe K, Suzuki N, Aoki M, Kawata A, Hirai T, Kato T et al

Tar DNA binding protein-43 (TDP-43) associates with stress granules:

(2010) Mutations of optineurin in amyotrophic lateral sclerosis. Nature

analysis of cultured cells and pathological brain tissue. PLoS ONE 5: e13250 Lomen-Hoerth C, Anderson T, Miller B (2002) The overlap of amyotrophic lateral sclerosis and frontotemporal dementia. Neurology 59: 1077 – 1079 Lopez-Gonzalez R, Lu Y, Gendron TF, Karydas A, Tran H, Yang D, Petrucelli L, Miller BL, Almeida S, Gao FB (2016) Poly(GR) in C9ORF72-related ALS/FTD compromises mitochondrial function and increases oxidative stress and DNA damage in iPSC-derived motor neurons. Neuron 92: 383 – 391 Lu Y, Zhang Z, Sun D, Sweeney ST, Gao FB (2013) Syntaxin 13, a genetic

465: 223 – 236 Mastrocola AS, Kim SH, Trinh AT, Rodenkirch LA, Tibbetts RS (2013) The RNAbinding protein fused in sarcoma (FUS) functions downstream of poly (ADP-ribose) polymerase (PARP) in response to DNA damage. J Biol Chem 288: 24731 – 24741 Matsumoto G, Shimogori T, Hattori N, Nukina N (2015) TBK1 controls autophagosomal engulfment of polyubiquitinated mitochondria through p62/SQSTM1 phosphorylation. Hum Mol Genet 24: 4429 – 4442 McDonald KK, Aulas A, Destroismaisons L, Pickles S, Beleac E, Camu W, Rouleau GA, Vande Velde C (2011) TAR DNA-binding protein 43 (TDP-43)

modifier of mutant CHMP2B in frontotemporal dementia, is required for

regulates stress granule dynamics via differential regulation of G3BP and

autophagosome maturation. Mol Cell 52: 264 – 271

TIA-1. Hum Mol Genet 20: 1400 – 1410

ª 2017 The Authors

The EMBO Journal

15

Published online: September 15, 2017

The EMBO Journal

Menzies FM, Fleming A, Caricasole A, Bento CF, Andrews SP, Ashkenazi A, Fullgrabe J, Jackson A, Jimenez Sanchez M, Karabiyik C, Licitra F, Lopez Ramirez A, Pavel M, Puri C, Renna M, Ricketts T, Schlotawa L, Vicinanza

Fen-Biao Gao et al

N’Diaye EN, Kajihara KK, Hsieh I, Morisaki H, Debnath J, Brown EJ (2009) PLIC proteins or ubiquilins regulate autophagy-dependent cell survival during nutrient starvation. EMBO Rep 10: 173 – 179

M, Won H, Zhu Y et al (2017) Autophagy and neurodegeneration:

Neary D, Snowden JS, Gustafson L, Passant U, Stuss D, Black S, Freedman M,

pathogenic mechanisms and therapeutic opportunities. Neuron 93:

Kertesz A, Robert PH, Albert M, Boone K, Miller BL, Cummings J, Benson

1015 – 1034

DF (1998) Frontotemporal lobar degeneration: a consensus on clinical

Meyer H, Bug M, Bremer S (2012) Emerging functions of the VCP/p97 AAAATPase in the ubiquitin system. Nat Cell Biol 14: 117 – 123 Mizielinska S, Lashley T, Norona FE, Clayton EL, Ridler CE, Fratta P, Isaacs AM (2013) C9orf72 frontotemporal lobar degeneration is characterised by

diagnostic criteria. Neurology 51: 1546 – 1554 Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, Bruce J, Schuck T, Grossman M, Clark CM, McCluskey LF, Miller BL, Masliah E, Mackenzie IR, Feldman H, Feiden W, Kretzschmar HA,

frequent neuronal sense and antisense RNA foci. Acta Neuropathol 126:

Trojanowski JQ, Lee VM (2006) Ubiquitinated TDP-43 in frontotemporal

845 – 857

lobar degeneration and amyotrophic lateral sclerosis. Science 314:

Mizielinska S, Gronke S, Niccoli T, Ridler CE, Clayton EL, Devoy A, Moens T, Norona FE, Woollacott IO, Pietrzyk J, Cleverley K, Nicoll AJ, Pickering-

130 – 133 Neumann M, Rademakers R, Roeber S, Baker M, Kretzschmar HA, Mackenzie

Brown S, Dols J, Cabecinha M, Hendrich O, Fratta P, Fisher EM,

IR (2009) A new subtype of frontotemporal lobar degeneration with FUS

Partridge L, Isaacs AM (2014) C9orf72 repeat expansions cause

pathology. Brain 132: 2922 – 2931

neurodegeneration in Drosophila through arginine-rich proteins. Science 345: 1192 – 1194 Moens TG, Partridge L, Isaacs AM (2017) Genetic models of C9orf72: what is toxic? Curr Opin Genet Dev 44: 92 – 101 Mohan A, Goodwin M, Swanson MS (2014) RNA-protein interactions in unstable microsatellite diseases. Brain Res 1584: 3 – 14 Molliex A, Temirov J, Lee J, Coughlin M, Kanagaraj AP, Kim HJ, Mittag T,

Neumann M, Valori CF, Ansorge O, Kretzschmar HA, Munoz DG, Kusaka H, Yokota O, Ishihara K, Ang LC, Bilbao JM, Mackenzie IR (2012) Transportin 1 accumulates specifically with FET proteins but no other transportin cargos in FTLD-FUS and is absent in FUS inclusions in ALS with FUS mutations. Acta Neuropathol 124: 705 – 716 Nishimura AL, Zupunski V, Troakes C, Kathe C, Fratta P, Howell M, Gallo JM, Hortobagyi T, Shaw CE, Rogelj B (2010) Nuclear import impairment causes

Taylor JP (2015) Phase separation by low complexity domains promotes

cytoplasmic trans-activation response DNA-binding protein accumulation

stress granule assembly and drives pathological fibrillization. Cell 163:

and is associated with frontotemporal lobar degeneration. Brain 133:

123 – 133 Mori K, Arzberger T, Grasser FA, Gijselinck I, May S, Rentzsch K, Weng SM, Schludi MH, van der Zee J, Cruts M, Van Broeckhoven C, Kremmer E, Kretzschmar HA, Haass C, Edbauer D (2013a) Bidirectional transcripts of the expanded C9orf72 hexanucleotide repeat are translated into aggregating dipeptide repeat proteins. Acta Neuropathol 126: 881 – 893 Mori K, Lammich S, Mackenzie IR, Forne I, Zilow S, Kretzschmar H, Edbauer

1763 – 1771 Nixon RA (2013) The role of autophagy in neurodegenerative disease. Nat Med 19: 983 – 997 Oakes JA, Davies MC, Collins MO (2017) TBK1: a new player in ALS linking autophagy and neuroinflammation. Mol Brain 10: 5 O’Rourke JG, Bogdanik L, Muhammad AK, Gendron TF, Kim KJ, Austin A, Cady J, Liu EY, Zarrow J, Grant S, Ho R, Bell S, Carmona S, Simpkinson M, Lall D,

D, Janssens J, Kleinberger G, Cruts M, Herms J, Neumann M, Van

Wu K, Daughrity L, Dickson DW, Harms MB, Petrucelli L et al (2015)

Broeckhoven C, Arzberger T, Haass C (2013b) hnRNP A3 binds to GGGGCC

C9orf72 BAC transgenic mice display typical pathologic features of ALS/

repeats and is a constituent of p62-positive/TDP43-negative inclusions in the hippocampus of patients with C9orf72 mutations. Acta Neuropathol 125: 413 – 423 Mori K, Weng SM, Arzberger T, May S, Rentzsch K, Kremmer E, Schmid B, Kretzschmar HA, Cruts M, Van Broeckhoven C, Haass C, Edbauer D (2013c) The C9orf72 GGGGCC repeat is translated into aggregating dipeptiderepeat proteins in FTLD/ALS. Science 339: 1335 – 1338 Morita M, Al-Chalabi A, Andersen PM, Hosler B, Sapp P, Englund E, Mitchell JE, Habgood JJ, de Belleroche J, Xi J, Jongjaroenprasert W, Horvitz HR,

FTD. Neuron 88: 892 – 901 O’Rourke JG, Bogdanik L, Yanez A, Lall D, Wolf AJ, Muhammad AK, Ho R, Carmona S, Vit JP, Zarrow J, Kim KJ, Bell S, Harms MB, Miller TM, Dangler CA, Underhill DM, Goodridge HS, Lutz CM, Baloh RH (2016) C9orf72 is required for proper macrophage and microglial function in mice. Science 351: 1324 – 1329 Panas MD, Ivanov P, Anderson P (2016) Mechanistic insights into mammalian stress granule dynamics. J Cell Biol 215: 313 – 323 Papadopoulos C, Kirchner P, Bug M, Grum D, Koerver L, Schulze N, Poehler

Gunnarsson LG, Brown RH Jr (2006) A locus on chromosome 9p confers

R, Dressler A, Fengler S, Arhzaouy K, Lux V, Ehrmann M, Weihl CC,

susceptibility to ALS and frontotemporal dementia. Neurology 66:

Meyer H (2017) VCP/p97 cooperates with YOD1, UBXD1 and PLAA to

839 – 844

drive clearance of ruptured lysosomes by autophagy. EMBO J 36:

Moscat J, Karin M, Diaz-Meco MT (2016) p62 in cancer: signaling adaptor beyond autophagy. Cell 167: 606 – 609 Murakami T, Qamar S, Lin JQ, Schierle GS, Rees E, Miyashita A, Costa AR,

135 – 150 Parkinson N, Ince PG, Smith MO, Highley R, Skibinski G, Andersen PM, Morrison KE, Pall HS, Hardiman O, Collinge J, Shaw PJ, Fisher EM, Study

Dodd RB, Chan FT, Michel CH, Kronenberg-Versteeg D, Li Y, Yang SP,

MRCPiA, Consortium FR (2006) ALS phenotypes with mutations in

Wakutani Y, Meadows W, Ferry RR, Dong L, Tartaglia GG, Favrin G, Lin WL

CHMP2B (charged multivesicular body protein 2B). Neurology 67:

et al (2015) ALS/FTD mutation-induced phase transition of FUS liquid droplets and reversible hydrogels into irreversible hydrogels impairs RNP granule function. Neuron 88: 678 – 690 Narayanan RK, Mangelsdorf M, Panwar A, Butler TJ, Noakes PG, Wallace RH (2013) Identification of RNA bound to the TDP-43 ribonucleoprotein

16

Pathogenic mechanisms in ALS and FTD

1074 – 1077 Passoni M, De Conti L, Baralle M, Buratti E (2012) UG repeats/TDP-43 interactions near 50 splice sites exert unpredictable effects on splicing modulation. J Mol Biol 415: 46 – 60 Patel A, Lee HO, Jawerth L, Maharana S, Jahnel M, Hein MY, Stoynov S,

complex in the adult mouse brain. Amyotroph Lateral Scler Frontotemporal

Mahamid J, Saha S, Franzmann TM, Pozniakovski A, Poser I, Maghelli N,

Degener 14: 252 – 260

Royer LA, Weigert M, Myers EW, Grill S, Drechsel D, Hyman AA, Alberti S

The EMBO Journal

ª 2017 The Authors

Published online: September 15, 2017

Fen-Biao Gao et al

The EMBO Journal

Pathogenic mechanisms in ALS and FTD

(2015) A liquid-to-solid phase transition of the ALS protein FUS accelerated by disease mutation. Cell 162: 1066 – 1077 Pesiridis GS, Tripathy K, Tanik S, Trojanowski JQ, Lee VM (2011) A “two-hit”

van Rheenen W, Shatunov A, Dekker AM, McLaughlin RL, Diekstra FP, Pulit SL, van der Spek RA, Vosa U, de Jong S, Robinson MR, Yang J, Fogh I, van Doormaal PT, Tazelaar GH, Koppers M, Blokhuis AM, Sproviero W, Jones

hypothesis for inclusion formation by carboxyl-terminal fragments of TDP-

AR, Kenna KP, van Eijk KR et al (2016) Genome-wide association analyses

43 protein linked to RNA depletion and impaired microtubule-dependent

identify new risk variants and the genetic architecture of amyotrophic

transport. J Biol Chem 286: 18845 – 18855 Peters OM, Cabrera GT, Tran H, Gendron TF, McKeon JE, Metterville J, Weiss A, Wightman N, Salameh J, Kim J, Sun H, Boylan KB, Dickson D, Kennedy

lateral sclerosis. Nat Genet 48: 1043 – 1048 Richter B, Sliter DA, Herhaus L, Stolz A, Wang C, Beli P, Zaffagnini G, Wild P, Martens S, Wagner SA, Youle RJ, Dikic I (2016) Phosphorylation of OPTN

Z, Lin Z, Zhang YJ, Daughrity L, Jung C, Gao FB, Sapp PC et al (2015)

by TBK1 enhances its binding to Ub chains and promotes selective

Human C9ORF72 hexanucleotide expansion reproduces RNA foci and

autophagy of damaged mitochondria. Proc Natl Acad Sci USA 113:

dipeptide repeat proteins but not neurodegeneration in BAC transgenic mice. Neuron 88: 902 – 909 Polymenidou M, Lagier-Tourenne C, Hutt KR, Huelga SC, Moran J, Liang TY, Ling SC, Sun E, Wancewicz E, Mazur C, Kordasiewicz H, Sedaghat Y, Donohue JP, Shiue L, Bennett CF, Yeo GW, Cleveland DW (2011) Long premRNA depletion and RNA missplicing contribute to neuronal vulnerability from loss of TDP-43. Nat Neurosci 14: 459 – 468 Pottier C, Bieniek KF, Finch N, van de Vorst M, Baker M, Perkersen R, Brown

4039 – 4044 Rogelj B, Easton LE, Bogu GK, Stanton LW, Rot G, Curk T, Zupan B, Sugimoto Y, Modic M, Haberman N, Tollervey J, Fujii R, Takumi T, Shaw CE, Ule J (2012) Widespread binding of FUS along nascent RNA regulates alternative splicing in the brain. Sci Rep 2: 603 Rossi S, Serrano A, Gerbino V, Giorgi A, Di Francesco L, Nencini M, Bozzo F, Schinina ME, Bagni C, Cestra G, Carri MT, Achsel T, Cozzolino M (2015) Nuclear accumulation of mRNAs underlies G4C2-repeat-induced

P, Ravenscroft T, van Blitterswijk M, Nicholson AM, DeTure M, Knopman

translational repression in a cellular model of C9orf72 ALS. J Cell Sci 128:

DS, Josephs KA, Parisi JE, Petersen RC, Boylan KB, Boeve BF, Graff-Radford

1787 – 1799

NR, Veltman JA, Gilissen C et al (2015) Whole-genome sequencing reveals

Rothenberg C, Srinivasan D, Mah L, Kaushik S, Peterhoff CM, Ugolino J, Fang

important role for TBK1 and OPTN mutations in frontotemporal lobar

S, Cuervo AM, Nixon RA, Monteiro MJ (2010) Ubiquilin functions in

degeneration without motor neuron disease. Acta Neuropathol 130: 77 – 92

autophagy and is degraded by chaperone-mediated autophagy. Hum Mol

Prince M, Bryce R, Albanese E, Wimo A, Ribeiro W, Ferri CP (2013) The global prevalence of dementia: a systematic review and metaanalysis. Alzheimers Dement 9: 63 – 75 Protter DS, Parker R (2016) Principles and properties of stress granules. Trends Cell Biol 26: 668 – 679 Prudencio M, Belzil VV, Batra R, Ross CA, Gendron TF, Pregent LJ, Murray ME, Overstreet KK, Piazza-Johnston AE, Desaro P, Bieniek KF, DeTure M, Lee

Genet 19: 3219 – 3232 Rubino E, Rainero I, Chio A, Rogaeva E, Galimberti D, Fenoglio P, Grinberg Y, Isaia G, Calvo A, Gentile S, Bruni AC, St George-Hyslop PH, Scarpini E, Gallone S, Pinessi L, Group TS (2012) SQSTM1 mutations in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Neurology 79: 1556 – 1562 Rulten SL, Rotheray A, Green RL, Grundy GJ, Moore DA, Gomez-Herreros F,

WC, Biendarra SM, Davis MD, Baker MC, Perkerson RB, van Blitterswijk M,

Hafezparast M, Caldecott KW (2014) PARP-1 dependent recruitment of the

Stetler CT, Rademakers R et al (2015) Distinct brain transcriptome profiles

amyotrophic lateral sclerosis-associated protein FUS/TLS to sites of

in C9orf72-associated and sporadic ALS. Nat Neurosci 18: 1175 – 1182 Qiu H, Lee S, Shang Y, Wang WY, Au KF, Kamiya S, Barmada SJ, Finkbeiner S, Lui H, Carlton CE, Tang AA, Oldham MC, Wang H, Shorter J, Filiano AJ, Roberson ED, Tourtellotte WG, Chen B, Tsai LH, Huang EJ (2014) ALSassociated mutation FUS-R521C causes DNA damage and RNA splicing defects. J Clin Invest 124: 981 – 999 Ramaswami M, Taylor JP, Parker R (2013) Altered ribostasis: RNA-protein granules in degenerative disorders. Cell 154: 727 – 736 Rauhut R, Fabrizio P, Dybkov O, Hartmuth K, Pena V, Chari A, Kumar V, Lee

oxidative DNA damage. Nucleic Acids Res 42: 307 – 314 Rulten SL, Grundy GJ (2017) Non-homologous end joining: common interaction sites and exchange of multiple factors in the DNA repair process. BioEssays 39: 1600209 Rusten TE, Vaccari T, Lindmo K, Rodahl LM, Nezis IP, Sem-Jacobsen C, Wendler F, Vincent JP, Brech A, Bilder D, Stenmark H (2007) ESCRTs and Fab1 regulate distinct steps of autophagy. Curr Biol 17: 1817 – 1825 Rutherford NJ, Zhang YJ, Baker M, Gass JM, Finch NA, Xu YF, Stewart H, Kelley

CT, Urlaub H, Kastner B, Stark H, Luhrmann R (2016) Molecular

BJ, Kuntz K, Crook RJ, Sreedharan J, Vance C, Sorenson E, Lippa C, Bigio EH,

architecture of the Saccharomyces cerevisiae activated spliceosome.

Geschwind DH, Knopman DS, Mitsumoto H, Petersen RC, Cashman NR

Science 353: 1399 – 1405 Reber S, Stettler J, Filosa G, Colombo M, Jutzi D, Lenzken SC, Schweingruber C, Bruggmann R, Bachi A, Barabino SM, Muhlemann O, Ruepp MD (2016) Minor intron splicing is regulated by FUS and affected by ALS-associated FUS mutants. EMBO J 35: 1504 – 1521 Reddy K, Zamiri B, Stanley SY, Macgregor RB Jr, Pearson CE (2013) The

et al (2008) Novel mutations in TARDBP (TDP-43) in patients with familial amyotrophic lateral sclerosis. PLoS Genet 4: e1000193 Sancar A, Lindsey-Boltz LA, Unsal-Kacmaz K, Linn S (2004) Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints. Annu Rev Biochem 73: 39 – 85 Sareen D, O’Rourke JG, Meera P, Muhammad AK, Grant S, Simpkinson M, Bell

disease-associated r(GGGGCC)n repeat from the C9orf72 gene forms tract

S, Carmona S, Ornelas L, Sahabian A, Gendron T, Petrucelli L, Baughn M,

length-dependent uni- and multimolecular RNA G-quadruplex structures. J

Ravits J, Harms MB, Rigo F, Bennett CF, Otis TS, Svendsen CN, Baloh RH

Biol Chem 288: 9860 – 9866 Renton AE, Majounie E, Waite A, Simon-Sanchez J, Rollinson S, Gibbs JR, Schymick JC, Laaksovirta H, van Swieten JC, Myllykangas L, Kalimo H, Paetau A, Abramzon Y, Remes AM, Kaganovich A, Scholz SW, Duckworth J, Ding J, Harmer DW, Hernandez DG et al (2011) A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 72: 257 – 268

ª 2017 The Authors

(2013) Targeting RNA foci in iPSC-derived motor neurons from ALS patients with a C9ORF72 repeat expansion. Sci Transl Med 5: 208ra149 Saudou F, Finkbeiner S, Devys D, Greenberg ME (1998) Huntingtin acts in the nucleus to induce apoptosis but death does not correlate with the formation of intranuclear inclusions. Cell 95: 55 – 66 Savas JN, Toyama BH, Xu T, Yates JR III, Hetzer MW (2012) Extremely longlived nuclear pore proteins in the rat brain. Science 335: 942

The EMBO Journal

17

Published online: September 15, 2017

The EMBO Journal

Schludi MH, May S, Grasser FA, Rentzsch K, Kremmer E, Kupper C,

Fen-Biao Gao et al

Suberbielle E, Sanchez PE, Kravitz AV, Wang X, Ho K, Eilertson K, Devidze N,

Klopstock T, German Consortium for Frontotemporal Lobar

Kreitzer AC, Mucke L (2013) Physiologic brain activity causes DNA double-

Degeneration, Bavarian Brain Banking Alliance, Arzberger T, Edbauer D

strand breaks in neurons, with exacerbation by amyloid-beta. Nat

(2015) Distribution of dipeptide repeat proteins in cellular models and C9orf72 mutation cases suggests link to transcriptional silencing. Acta Neuropathol 130: 537 – 555 Sellier C, Campanari ML, Julie Corbier C, Gaucherot A, Kolb-Cheynel I, OuladAbdelghani M, Ruffenach F, Page A, Ciura S, Kabashi E, Charlet-

Neurosci 16: 613 – 621 Sullivan PM, Zhou X, Robins AM, Paushter DH, Kim D, Smolka MB, Hu F (2016) The ALS/FTLD associated protein C9orf72 associates with SMCR8 and WDR41 to regulate the autophagy-lysosome pathway. Acta Neuropathol Commun 4: 51

Berguerand N (2016) Loss of C9ORF72 impairs autophagy and synergizes

Sun Z, Diaz Z, Fang X, Hart MP, Chesi A, Shorter J, Gitler AD (2011) Molecular

with polyQ Ataxin-2 to induce motor neuron dysfunction and cell death.

determinants and genetic modifiers of aggregation and toxicity for the

EMBO J 35: 1276 – 1297 Sellier C, Buijsen RA, He F, Natla S, Jung L, Tropel P, Gaucherot A, Jacobs H,

ALS disease protein FUS/TLS. PLoS Biol 9: e1000614 Sun S, Ling SC, Qiu J, Albuquerque CP, Zhou Y, Tokunaga S, Li H, Qiu H, Bui A,

Meziane H, Vincent A, Champy MF, Sorg T, Pavlovic G, Wattenhofer-Donze

Yeo GW, Huang EJ, Eggan K, Zhou H, Fu XD, Lagier-Tourenne C, Cleveland

M, Birling MC, Oulad-Abdelghani M, Eberling P, Ruffenach F, Joint M,

DW (2015) ALS-causative mutations in FUS/TLS confer gain and loss of

Anheim M et al (2017) Translation of expanded CGG repeats into

function by altered association with SMN and U1-snRNP. Nat Commun 6:

FMRpolyG is pathogenic and may contribute to fragile X tremor ataxia syndrome. Neuron 93: 331 – 347 Sephton CF, Cenik C, Kucukural A, Dammer EB, Cenik B, Han Y, Dewey CM,

6171 Sun M, Bell W, LaClair KD, Ling JP, Han H, Kageyama Y, Pletnikova O, Troncoso JC, Wong PC, Chen LL (2017) Cryptic exon incorporation

Roth FP, Herz J, Peng J, Moore MJ, Yu G (2011) Identification of neuronal

occurs in Alzheimer’s brain lacking TDP-43 inclusion but

RNA targets of TDP-43-containing ribonucleoprotein complexes. J Biol

exhibiting nuclear clearance of TDP-43. Acta Neuropathol 133:

Chem 286: 1204 – 1215

923 – 931

Shang J, Yamashita T, Nakano Y, Morihara R, Li X, Feng T, Liu X, Huang Y,

Takeuchi R, Toyoshima Y, Tada M, Shiga A, Tanaka H, Shimohata M, Kimura

Fukui Y, Hishikawa N, Ohta Y, Abe K (2017) Aberrant distributions of

K, Morita T, Kakita A, Nishizawa M, Takahashi H (2013) Transportin 1

nuclear pore complex proteins in ALS mice and ALS patients. Neuroscience

accumulates in FUS inclusions in adult-onset ALS without FUS mutation.

350: 158 – 168 Shelkovnikova TA, Robinson HK, Connor-Robson N, Buchman VL (2013)

Neuropathol Appl Neurobiol 39: 580 – 584 Tan Q, Yalamanchili HK, Park J, De Maio A, Lu HC, Wan YW, White JJ, Bondar

Recruitment into stress granules prevents irreversible aggregation of FUS

VV, Sayegh LS, Liu X, Gao Y, Sillitoe RV, Orr HT, Liu Z, Zoghbi HY (2016)

protein mislocalized to the cytoplasm. Cell Cycle 12: 3194 – 3202

Extensive cryptic splicing upon loss of RBM17 and TDP43 in

Shen WC, Li HY, Chen GC, Chern Y, Tu PH (2015) Mutations in the ubiquitin-

neurodegeneration models. Hum Mol Genet 25: 5083 – 5093

binding domain of OPTN/optineurin interfere with autophagy-mediated

Tollervey JR, Curk T, Rogelj B, Briese M, Cereda M, Kayikci M, Konig J, Hortobagyi

degradation of misfolded proteins by a dominant-negative mechanism.

T, Nishimura AL, Zupunski V, Patani R, Chandran S, Rot G, Zupan B, Shaw CE,

Autophagy 11: 685 – 700

Ule J (2011) Characterizing the RNA targets and position-dependent splicing

Shi KY, Mori E, Nizami ZF, Lin Y, Kato M, Xiang S, Wu LC, Ding M, Yu Y, Gall JG, McKnight SL (2017) Toxic PRn poly-dipeptides encoded by the C9orf72

regulation by TDP-43. Nat Neurosci 14: 452 – 458 Toyama BH, Savas JN, Park SK, Harris MS, Ingolia NT, Yates JR III, Hetzer MW

repeat expansion block nuclear import and export. Proc Natl Acad Sci USA

(2013) Identification of long-lived proteins reveals exceptional stability of

114: E1111 – E1117

essential cellular structures. Cell 154: 971 – 982

Singh RK, Cooper TA (2012) Pre-mRNA splicing in disease and therapeutics. Trends Mol Med 18: 472 – 482 Skibinski G, Parkinson NJ, Brown JM, Chakrabarti L, Lloyd SL, Hummerich H, Nielsen JE, Hodges JR, Spillantini MG, Thusgaard T, Brandner S, Brun A, Rossor MN, Gade A, Johannsen P, Sorensen SA, Gydesen S, Fisher EM,

Tran H, Almeida S, Moore J, Gendron TF, Chalasani U, Lu Y, Du X, Nickerson JA, Petrucelli L, Weng Z, Gao FB (2015) Differential toxicity of nuclear RNA foci versus dipeptide repeat proteins in a Drosophila model of C9ORF72 FTD/ALS. Neuron 87: 1207 – 1214 Tresse E, Salomons FA, Vesa J, Bott LC, Kimonis V, Yao TP, Dantuma NP,

Collinge J (2005) Mutations in the endosomal ESCRTIII-complex

Taylor JP (2010) VCP/p97 is essential for maturation of ubiquitin-

subunit CHMP2B in frontotemporal dementia. Nat Genet 37:

containing autophagosomes and this function is impaired by mutations

806 – 808 Skourti-Stathaki K, Proudfoot NJ (2014) A double-edged sword: R loops as

that cause IBMPFD. Autophagy 6: 217 – 227 Troakes C, Hortobagyi T, Vance C, Al-Sarraj S, Rogelj B, Shaw CE (2013)

threats to genome integrity and powerful regulators of gene expression.

Transportin 1 colocalization with Fused in Sarcoma (FUS) inclusions is not

Genes Dev 28: 1384 – 1396

characteristic for amyotrophic lateral sclerosis-FUS confirming disrupted

Sreedharan J, Blair IP, Tripathi VB, Hu X, Vance C, Rogelj B, Ackerley S, Durnall JC, Williams KL, Buratti E, Baralle F, de Belleroche J, Mitchell JD, Leigh PN, Al-Chalabi A, Miller CC, Nicholson G, Shaw CE (2008) TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science 319: 1668 – 1672 Stalekar M, Yin X, Rebolj K, Darovic S, Troakes C, Mayr M, Shaw CE, Rogelj B (2015) Proteomic analyses reveal that loss of TDP-43 affects RNA

18

Pathogenic mechanisms in ALS and FTD

nuclear import of mutant FUS and distinguishing it from frontotemporal lobar degeneration with FUS inclusions. Neuropathol Appl Neurobiol 39: 553 – 561 Ugolino J, Ji YJ, Conchina K, Chu J, Nirujogi RS, Pandey A, Brady NR, Hamacher-Brady A, Wang J (2016) Loss of C9orf72 enhances autophagic activity via deregulated mTOR and TFEB signaling. PLoS Genet 12: e1006443

processing and intracellular transport. Neuroscience 293: 157 – 170

Vance C, Al-Chalabi A, Ruddy D, Smith BN, Hu X, Sreedharan J, Siddique T,

Strong MJ, Lomen-Hoerth C, Caselli RJ, Bigio EH, Yang W (2003) Cognitive

Schelhaas HJ, Kusters B, Troost D, Baas F, de Jong V, Shaw CE (2006)

impairment, frontotemporal dementia, and the motor neuron diseases.

Familial amyotrophic lateral sclerosis with frontotemporal dementia is

Ann Neurol 54(Suppl 5): S20 – S23

linked to a locus on chromosome 9p13.2-21.3. Brain 129: 868 – 876

The EMBO Journal

ª 2017 The Authors

Published online: September 15, 2017

Fen-Biao Gao et al

The EMBO Journal

Pathogenic mechanisms in ALS and FTD

Vance C, Rogelj B, Hortobagyi T, De Vos KJ, Nishimura AL, Sreedharan J,

Meyer K, Shaw PJ, Grierson AJ, De Vos KJ (2016) The C9orf72 protein

Hu X, Smith B, Ruddy D, Wright P, Ganesalingam J, Williams KL,

interacts with Rab1a and the ULK1 complex to regulate initiation of

Tripathi V, Al-Saraj S, Al-Chalabi A, Leigh PN, Blair IP, Nicholson G, de Belleroche J, Gallo JM et al (2009) Mutations in FUS, an RNA processing

autophagy. EMBO J 35: 1656 – 1676 Wen X, Tan W, Westergard T, Krishnamurthy K, Markandaiah SS, Shi Y, Lin S,

protein, cause familial amyotrophic lateral sclerosis type 6. Science 323:

Shneider NA, Monaghan J, Pandey UB, Pasinelli P, Ichida JK, Trotti D

1208 – 1211

(2014) Antisense proline-arginine RAN dipeptides linked to C9ORF72-ALS/

Vance C, Scotter EL, Nishimura AL, Troakes C, Mitchell JC, Kathe C, Urwin H, Manser C, Miller CC, Hortobagyi T, Dragunow M, Rogelj B, Shaw CE (2013) ALS mutant FUS disrupts nuclear localization and sequesters

FTD form toxic nuclear aggregates that initiate in vitro and in vivo neuronal death. Neuron 84: 1213 – 1225 Williams KL, Topp S, Yang S, Smith B, Fifita JA, Warraich ST, Zhang KY,

wild-type FUS within cytoplasmic stress granules. Hum Mol Genet 22:

Farrawell N, Vance C, Hu X, Chesi A, Leblond CS, Lee A, Rayner SL,

2676 – 2688

Sundaramoorthy V, Dobson-Stone C, Molloy MP, van Blitterswijk M,

Vatsavayai SC, Yoon SJ, Gardner RC, Gendron TF, Vargas JN, Trujillo A, Pribadi M, Phillips JJ, Gaus SE, Hixson JD, Garcia PA, Rabinovici GD, Coppola G, Geschwind DH, Petrucelli L, Miller BL, Seeley WW (2016) Timing and significance of pathological features in C9orf72 expansion-associated frontotemporal dementia. Brain 139: 3202 – 3216 Vercelletto M, Ronin M, Huvet M, Magne C, Feve JR (1999) Frontal type

Dickson DW, Petersen RC et al (2016) CCNF mutations in amyotrophic lateral sclerosis and frontotemporal dementia. Nat Commun 7: 11253 Wimo A, Guerchet M, Ali GC, Wu YT, Prina AM, Winblad B, Jonsson L, Liu Z, Prince M (2017) The worldwide costs of dementia 2015 and comparisons with 2010. Alzheimers Dement 13: 1 – 7 Winton MJ, Igaz LM, Wong MM, Kwong LK, Trojanowski JQ, Lee VM (2008)

dementia preceding amyotrophic lateral sclerosis: a neuropsychological

Disturbance of nuclear and cytoplasmic TAR DNA-binding protein (TDP-43)

and SPECT study of five clinical cases. Eur J Neurol 6: 295 – 299

induces disease-like redistribution, sequestration, and aggregate

Vernay A, Therreau L, Blot B, Risson V, Dirrig-Grosch S, Waegaert R, Lequeu T, Sellal F, Schaeffer L, Sadoul R, Loeffler JP, Rene F (2016) A transgenic

formation. J Biol Chem 283: 13302 – 13309 Woerner AC, Frottin F, Hornburg D, Feng LR, Meissner F, Patra M, Tatzelt J,

mouse expressing CHMP2Bintron5 mutant in neurons develops

Mann M, Winklhofer KF, Hartl FU, Hipp MS (2016) Cytoplasmic protein

histological and behavioural features of amyotrophic lateral sclerosis and

aggregates interfere with nucleocytoplasmic transport of protein and RNA.

frontotemporal dementia. Hum Mol Genet 25: 3341 – 3360 Wagner MJ, Kim TH, Savall J, Schnitzer MJ, Luo L (2017) Cerebellar granule cells encode the expectation of reward. Nature 544: 96 – 100 Waite AJ, Baumer D, East S, Neal J, Morris HR, Ansorge O, Blake DJ (2014)

Science 351: 173 – 176 Wolozin B (2012) Regulated protein aggregation: stress granules and neurodegeneration. Mol Neurodegener 7: 56 Wong YC, Holzbaur EL (2014) Optineurin is an autophagy receptor for

Reduced C9orf72 protein levels in frontal cortex of amyotrophic

damaged mitochondria in parkin-mediated mitophagy that is

lateral sclerosis and frontotemporal degeneration brain with the

disrupted by an ALS-linked mutation. Proc Natl Acad Sci USA 111:

C9ORF72 hexanucleotide repeat expansion. Neurobiol Aging 35: 1779.e5 – 1779.e13 Walker C, Herranz-Martin S, Karyka E, Liao C, Lewis K, Elsayed W, Lukashchuk V, Chiang SC, Ray S, Mulcahy PJ, Jurga M, Tsagakis I, Iannitti T, Chandran J, Coldicott I, De VK, Hassan MK, Higginbottom A, Shaw PJ, Hautbergue

E4439 – E4448 Xiao S, Sanelli T, Dib S, Sheps D, Findlater J, Bilbao J, Keith J, Zinman L, Rogaeva E, Robertson J (2011) RNA targets of TDP-43 identified by UVCLIP are deregulated in ALS. Mol Cell Neurosci 47: 167 – 180 Xiao S, MacNair L, McGoldrick P, McKeever PM, McLean JR, Zhang M, Keith J,

GM et al (2017) C9orf72 expansion disrupts ATM-mediated chromosomal

Zinman L, Rogaeva E, Robertson J (2015) Isoform-specific antibodies reveal

break repair. Nat Neurosci 20: 1225 – 1235

distinct subcellular localizations of C9orf72 in amyotrophic lateral

Wang Y, Martinez-Vicente M, Krüger U, Kaushik S, Wong E, Mandelkow EM, Cuervo AM, Mandelkow E (2009) Tau fragmentation, aggregation and

sclerosis. Ann Neurol 78: 568 – 583 Xu Z, Poidevin M, Li X, Li Y, Shu L, Nelson DL, Li H, Hales CM, Gearing M,

clearance: the dual role of lysosomal processing. Hum Mol Genet 18:

Wingo TS, Jin P (2013) Expanded GGGGCC repeat RNA associated with

4153 – 4170

amyotrophic lateral sclerosis and frontotemporal dementia causes

Wang WY, Pan L, Su SC, Quinn EJ, Sasaki M, Jimenez JC, Mackenzie IR, Huang EJ, Tsai LH (2013) Interaction of FUS and HDAC1 regulates DNA damage response and repair in neurons. Nat Neurosci 16: 1383 – 1391 Wang G, Yang H, Yan S, Wang CE, Liu X, Zhao B, Ouyang Z, Yin P, Liu Z, Zhao Y, Liu T, Fan N, Guo L, Li S, Li XJ, Lai L (2015) Cytoplasmic mislocalization of RNA splicing factors and aberrant neuronal gene splicing in TDP-43 transgenic pig brain. Mol Neurodegener 10: 42 Ward ME, Taubes A, Chen R, Miller BL, Sephton CF, Gelfand JM, Minami S, Boscardin J, Martens LH, Seeley WW, Yu G, Herz J, Filiano AJ, Arrant AE, Roberson ED, Kraft TW, Farese RV Jr, Green A, Gan L (2014) Early retinal

neurodegeneration. Proc Natl Acad Sci USA 110: 7778 – 7783 Yan C, Wan R, Bai R, Huang G, Shi Y (2017) Structure of a yeast step II catalytically activated spliceosome. Science 355: 149 – 155 Yang L, Embree LJ, Tsai S, Hickstein DD (1998) Oncoprotein TLS interacts with serine-arginine proteins involved in RNA splicing. J Biol Chem 273: 27761 – 27764 Yang D, Abdallah A, Li Z, Lu Y, Almeida S, Gao FB (2015) FTD/ALS-associated poly(GR) protein impairs the Notch pathway and is recruited by poly(GA) into cytoplasmic inclusions. Acta Neuropathol 130: 525 – 535 Yang M, Liang C, Swaminathan K, Herrlinger S, Lai F, Shiekhattar R, Chen JF

neurodegeneration and impaired Ran-mediated nuclear import of TDP-

(2016) A C9ORF72/SMCR8-containing complex regulates ULK1 and plays a

43 in progranulin-deficient FTLD. J Exp Med 211: 1937 – 1945

dual role in autophagy. Sci Adv 2: e1601167

Watts GD, Wymer J, Kovach MJ, Mehta SG, Mumm S, Darvish D, Pestronk A,

Yin S, Lopez-Gonzalez R, Kunz RC, Gangopadhyay J, Borufka C, Gygi SP, Gao

Whyte MP, Kimonis VE (2004) Inclusion body myopathy associated with

F-B, Reed R (2017) Evidence that C9ORF72 dipeptide repeat proteins

Paget disease of bone and frontotemporal dementia is caused by mutant

associate with U2 snRNP to cause mis-splicing in ALS/FTD patients. Cell

valosin-containing protein. Nat Genet 36: 377 – 381 Webster CP, Smith EF, Bauer CS, Moller A, Hautbergue GM, Ferraiuolo L, Myszczynska MA, Higginbottom A, Walsh MJ, Whitworth AJ, Kaspar BK,

ª 2017 The Authors

Rep 19: 2244 – 2256 Yokoseki A, Shiga A, Tan CF, Tagawa A, Kaneko H, Koyama A, Eguchi H, Tsujino A, Ikeuchi T, Kakita A, Okamoto K, Nishizawa M, Takahashi H,

The EMBO Journal

19

Published online: September 15, 2017

The EMBO Journal

Fen-Biao Gao et al

Onodera O (2008) TDP-43 mutation in familial amyotrophic lateral

Lloyd TE et al (2015) The C9orf72 repeat expansion disrupts

sclerosis. Ann Neurol 63: 538 – 542

nucleocytoplasmic transport. Nature 525: 56 – 61

Yu Y, Reed R (2015) FUS functions in coupling transcription to splicing by

Zhang YJ, Gendron TF, Grima JC, Sasaguri H, Jansen-West K, Xu YF,

mediating an interaction between RNAP II and U1 snRNP. Proc Natl Acad

Katzman RB, Gass J, Murray ME, Shinohara M, Lin WL, Garrett A,

Sci USA 112: 8608 – 8613

Stankowski JN, Daughrity L, Tong J, Perkerson EA, Yue M, Chew J,

Yu Y, Chi B, Xia W, Gangopadhyay J, Yamazaki T, Winkelbauer-Hurt ME, Yin S,

Castanedes-Casey M, Kurti A et al (2016) C9ORF72 poly(GA) aggregates

Eliasse Y, Adams E, Shaw CE, Reed R (2015) U1 snRNP is mislocalized in

sequester and impair HR23 and nucleocytoplasmic transport proteins.

ALS patient fibroblasts bearing NLS mutations in FUS and is required for motor neuron outgrowth in zebrafish. Nucleic Acids Res 43: 3208 – 3218 Zhang J, Ito H, Wate R, Ohnishi S, Nakano S, Kusaka H (2006) Altered

Nat Neurosci 19: 668 – 677 Zu T, Gibbens B, Doty NS, Gomes-Pereira M, Huguet A, Stone MD, Margolis J, Peterson M, Markowski TW, Ingram MA, Nan Z, Forster C,

distributions of nucleocytoplasmic transport-related proteins in the spinal

Low WC, Schoser B, Somia NV, Clark HB, Schmechel S, Bitterman PB,

cord of a mouse model of amyotrophic lateral sclerosis. Acta Neuropathol

Gourdon G, Swanson MS et al (2011) Non-ATG-initiated translation

112: 673 – 680

directed by microsatellite expansions. Proc Natl Acad Sci USA 108:

Zhang D, Iyer LM, He F, Aravind L (2012) Discovery of novel DENN proteins: implications for the evolution of eukaryotic intracellular membrane structures and human disease. Front Genet 3: 283 Zhang K, Donnelly CJ, Haeusler AR, Grima JC, Machamer JB, Steinwald P,

20

Pathogenic mechanisms in ALS and FTD

260 – 265 Zu T, Liu Y, Banez-Coronel M, Reid T, Pletnikova O, Lewis J, Miller TM, Harms MB, Falchook AE, Subramony SH, Ostrow LW, Rothstein JD, Troncoso JC, Ranum LP (2013) RAN proteins and RNA foci from antisense transcripts in

Daley EL, Miller SJ, Cunningham KM, Vidensky S, Gupta S, Thomas MA,

C9ORF72 ALS and frontotemporal dementia. Proc Natl Acad Sci USA 110:

Hong I, Chiu SL, Huganir RL, Ostrow LW, Matunis MJ, Wang J, Sattler R,

E4968 – E4977

The EMBO Journal

ª 2017 The Authors

Dysregulated molecular pathways in amyotrophic lateral sclerosis-frontotemporal dementia spectrum disorder.

Frontotemporal dementia (FTD), the second most common form of dementia in people under 65 years of age, is characterized by progressive atrophy of the...
928KB Sizes 0 Downloads 22 Views