Anda di halaman 1dari 13

Review

TDP-43 and FUS in amyotrophic lateral sclerosis and


frontotemporal dementia
Ian R A Mackenzie, Rosa Rademakers, Manuela Neumann

Abnormal intracellular protein aggregates comprise a key characteristic in most neurodegenerative diseases, including Lancet Neurol 2010; 9: 995–1007
amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). The seminal discoveries of accumulation of See In Context page 955
TDP-43 in most cases of ALS and the most frequent form of FTD, frontotemporal lobar degeneration with Department of Pathology and
ubiquitinated inclusions, followed by identification of FUS as the novel pathological protein in a small subset of Laboratory Medicine,
Vancouver General Hospital,
patients with ALS and various FTD subtypes provide clear evidence that these disorders are related. The creation of a
Vancouver, BC, Canada
novel molecular classification of ALS and FTD based on the identity of the predominant protein abnormality has, (Prof I R A Mackenzie MD);
therefore, been possible. The striking functional and structural similarities of TDP-43 and FUS, which are both Department of Neuroscience,
DNA/RNA binding proteins, imply that abnormal RNA metabolism is a pivotal event, but the mechanisms leading to Mayo Clinic College of
Medicine, Jacksonville, FL, USA
TDP-43 and FUS accumulation and the resulting neurodegeneration are currently unknown. Nonetheless, TDP-43
(R Rademakers PhD); and
and FUS are promising candidates for the development of novel biomarker assays and targeted therapies. Institute of Neuropathology,
University Hospital of Zurich,
Introduction patients with ALS due to SOD1 mutations (ALS-SOD), Zurich, Switzerland
(M Neumann MD)
The presence of abnormal protein aggregates in the but is not seen in inclusions in other ALS forms.7 This
Correspondence to:
cytoplasm of neurons is a key characteristic of most finding indicated that other pathological proteins
Dr Manuela Neumann, Institute
neurodegenerative diseases. In several common disorders remained to be discovered. of Neuropathology, University
the disease-specific pathological protein was identified FTD is the second most frequent cause of presenile Hospital of Zurich,
decades ago (eg, tau in neurofibrillary tangles of dementia and is a clinically, genetically, and Schmelzbergstrasse 12,
8091 Zurich, Switzerland
Alzheimer’s disease and α-synuclein in Lewy bodies of pathologically heterogeneous syndrome. The clinical
manuela.neumann@usz.ch
Parkinson’s disease), providing the logical basis for phenotypes include behavioural variant FTD, semantic
research into the underlying biological mechanisms, dementia, and progressive non-fluent aphasia.8 The
diagnosis, and treatment. The biochemical composition associated neuropathology of all phenotypes is
of the ubiquitinated inclusions in most cases of characterised by FTLD, but the microscopic features
amyotrophic lateral sclerosis (ALS) and the most frequent vary. Historically, FTLD subtypes were classified as
pathological form of frontotemporal dementia (FTD), those associated with abnormal accumulation of tau
frontotemporal lobar degeneration with ubiquitinated (FTLD-tau) and those with tau-negative, ubiquitin-
inclusions (FTLD-U), remained unknown until the TAR positive inclusions (FTLD-U).8 The observation that
DNA binding protein 43 (TDP-43)1 and the fused in some ALS patients develop cognitive deficits with
sarcoma protein ([FUS] also known as translocated in prominent frontal lobe features and that in those
liposarcoma)2,3 were identified as disease-related proteins patients the associated neuropathology resembles
in these two disorders. This Review highlights the novel FTLD-U led to the idea that ALS and FTD with FTLD-U
molecular classification of ALS and FTLD that emerged might be related.9–11
from these findings and summarises developments This hypothesis was confirmed in 2006 with the
in the clinicopathological and genetic knowledge of identification of the DNA/RNA binding protein TDP-43
disorders involving TDP-43 and FUS. Particular as the pathological protein in most cases of FTLD-U,1,12
emphasis is placed on the molecular features and (renamed as FTLD-TDP)13 and SOD1-negative ALS
their pathomechanistic implications. (ALS-TDP).1,14 This discovery prompted researchers to
prioritise genetic analysis of functionally related genes
Novel molecular classification in the chromosome 16 region that had previously been
ALS is the most frequent form of motor neuron disease, linked to some familial forms of ALS, and led to
in which the loss of motor neurons from the brain and the identification of mutations in FUS as the genetic
spinal cord leads to fatal paralysis and death, generally cause, and of FUS as the pathological protein in
within 1–5 years.4 A characteristic feature of degenerating TDP-43-negative, SOD1-negative ALS (ALS-FUS).2,3
neurons is the presence of cytoplasmic inclusions FUS was subsequently identified as the pathological
positive for ubiquitin. Approximately 5% of patients with protein in most of the remaining tau-negative,
ALS have a positive family history of the disorder. The TDP-43-negative FTLD subtypes (renamed as FTLD-
first pathological mutations identified in ALS were in FUS).13 A novel molecular classification was, therefore,
SOD1,5 and account for around 20% of familial ALS created probably reflecting distinct underlying
cases. That discovery has been the basis for most ALS pathological mechanisms in the different subtypes
research in the past decade, which has provided (figure 1).15–21 Moreover, the data provided clear molecular
important insights into SOD1-mediated neurotoxic evidence that ALS and FTD represent a clinico-
effects.6 Misfolded SOD1 is present in the inclusions in pathological spectrum of diseases.

www.thelancet.com/neurology Vol 9 October 2010 995


Review

Ubiquitin-positive inclusions in FTLD-U and ALS

Disease
TDP-43 FUS SOD1 Unknown
protein

FTLD-TDP types 1–4 ALS-TDP FTLD-FUS ALS-FUS ALS-SOD FTLD-UPS


Clinico-
pathological Unclassifiable Atypical FTLD-U
subtypes NIFID
BIBD

Associated GRN TARDBP (FUS)‡ FUS SOD1 CHMP2B


genes Chromosome 9p (ANG)†
VCP
(TARDBP)*

Figure 1: New molecular classification of FTLD-U and ALS


Both disorders are characterised by ubiquitin-positive inclusions. The ubiquitinated protein in most cases of FTLD-U and ALS is TDP-43, and these cases should be
classified as FTLD-TDP and ALS-TDP, respectively. In a small subset of cases of FTLD-U and ALS, the pathological protein is FUS, and these should be classified as
FTLD-FUS and ALS-FUS, respectively. Misfolded SOD1 is the pathological protein in ubiquitin-positive inclusions in ALS with SOD1 mutations, and these are classified
as ALS-SOD. In a few cases of FTLD-U, the ubiquitinated protein is unknown, and these cases are classified as FTLD-UPS, to indicate that inclusions are currently
labelled only by markers of the ubiquitin-proteasome system. ALS=amyotrophic lateral sclerosis. BIBD=basophilic inclusion body disease. FTD=frontotemporal
dementia. FTLD=frontotemporal lobar degeneration. FTLD-U=FTLD with ubiquitinated inclusions. NIFID=neuronal intermediate filament inclusion disease.
UPS=ubiquitin-proteasome system. *Rare reports of TARDBP mutations in patients with FTD or ALS with FTD.15–17 †One case report.18 ‡Rare reports of FUS mutations
in patients with FTD or ALS with FTD.19–21

R495X R514G/S R521G/C/H and a Gly-rich C-terminal region that allow it to bind
R216C R234C/L G507D R514S;G515C R522G single-stranded DNA, RNA, and proteins.22,23 The exon
G225V R244C K510R/E H517Q*/P R524W/T/S skipping and splicing inhibitory activity requires the
G230C M254V S513P R518G/K P525L
C-terminal region, which interacts with other members
G187S S462F
S57del G156E G191S S402_P411delinsGGGG G466VfsX14 of the heterogeneous nuclear ribonucleoprotein family.24
TDP-43 can continuously shuttle between the nucleus
FUS QGSY-rich region Gly-rich RRM RGG-rich ZnF RGG-rich 526 aa
and cytoplasm—a process partly regulated by nuclear
NES NLS
localisation signal and nuclear export signal motifs.25,26
NLS NES
In addition to the well characterised role of TDP-43 in
TDP-43 RRM1 RRM2 Gly-rich 414 aa
transcription and splicing regulation, roles have been
D169G K263E suggested in other cellular processes, such as microRNA
N267S processing, apoptosis, cell division, and stabilisation of
G287S G294A/V M311V Q343R Y374X I383V messenger RNA.27–31 Early embryonic death in TDP-43
G290A G295R/S A315T N345K N378D G384R
S292N G298S A321V/G G348C/V S379P/C N390D/S knockout mice supports an essential role for this protein
Q331K N352T/S A382P/T S393L in normal development.32–34 In the brain, transient
S332N R361S
G335D P363A
redistribution to the cytoplasm could be a normal
M337V response to neuronal injury.35,36 Moreover, studies have
suggested a role in the regulation of neuronal plasticity37
Figure 2: Schematic overview of protein domains of FUS and TDP-43 and identified gene mutations
and maintenance of dendritic integrity.38
associated with ALS and FTLD
Mutations identified so far in FUS and TARDBP are summarised by their predicted protein changes and shown with
their relative positions in the protein domains of FUS (Genbank Accession number CAG33028.1) and TDP-43 Clinicopathological spectrum of TDP-43 proteinopathies
(Genbank Accession number NP_031401.1). Mutations shown in black were identified in patients with ALS only, TDP-43 proteinopathies are defined by the presence of
and mutations in blue were also associated with FTD phenotypes. ALS=amyotrophic lateral sclerosis.
inclusions composed of abnormal TDP-43. Antibodies
FTLD=frontotemporal lobar degeneration. NES=nuclear export signal. NLS=nuclear localisation signal.
QGSY=Gln-Gly-Ser-Tyr-rich region. RGG=Arg-Gly-Gly-rich motif. RRM=RNA recognition motif. against TDP-43 consistently label the inclusions in
ZnF=Cys2/Cys2-type zinc finger motif. FTLD-TDP and ALS-TDP, but not those in various other
neurodegenerative disorders.1,12,39,40 They also label
TDP-43 previously unrecognised ubiquitin-negative pathologies,
Normal function including glial cytoplasmic inclusions,41 neurons that
TDP-43 is a 414 aminoacid protein encoded by TARDBP showed diffuse granular cytoplasmic staining (so-called
on chromosome 1p36.2, and consists of five coding and preinclusions),42 and delicate neurites in the CA1 region.43
one non-coding exon (figure 2). The protein is highly Of potential functional importance, the presence of
conserved, widely expressed, and predominantly localised TDP-43-positive inclusions is consistently associated
to the nucleus.22 It contains two RNA recognition motifs with a substantial reduction in normal physiological

996 www.thelancet.com/neurology Vol 9 October 2010


Review

A B I

Control FTLD-TDP FTLD-TDP

100

C D 75 ***

MW (kDa)
50
**

E F
37

25 *

Pan TDP Phospho-


G H specific TDP

Figure 3: Pathological features in ALS-TDP and FTLD-TDP


TDP-43 immunohistochemistry on paraffin-embedded tissue showing (A) TDP-43-immunoreactive skein-like and (B) round inclusions in motor neurons in ALS-TDP.
FTLD-TDP pathology is subdivided into four distinct morphological subtypes: (C) type 1 is characterised by compact neuronal cytoplasmic inclusions and short
neurites; (D) type 2 is characterised by long neurites; (E) type 3 is characterised by compact and granular cytoplasmic inclusions; and (F) type 4 is characterised by
numerous neuronal intranuclear inclusions. (G) Cytoplasmic inclusions in the dentate granule cells of the hippocampus. Nuclear staining is absent in inclusion-bearing
cells. (H) Glial cytoplasmic inclusions. Scale bars: 40 μm (D), 20 μm (A, C, G), 15 μm (E, F), and 10 μm (B, H). (I) Immunoblot analysis of urea fractions isolated from
brain tissue, showing the highly characteristic biochemical signature of TDP-43 in FTLD-TDP, with pathological bands of approximately 25 kDa (*) and 45 kDa (**), and
a high-molecular-weight smear (***) that are not detected in controls. The arrow indicates the wild-type 43 kDa TDP-43 band present in controls and in patients with
FTLD-TDP. Notably, only pathological TDP-43 is detected by a phosphorylation-specific TDP-43 antibody against phosphorylated serine residues 409 and 410.45
ALS=amyotrophic lateral sclerosis. ALS-TDP=TDP-43-positive ALS. FTLD-TDP=TDP-43-positive frontotemporal lobar degeneration.

diffuse nuclear staining.1,39 Pathological forms of TDP-43 filamentous skeins (figure 3). No obvious correlation
show evidence of abnormal processing, being hyper- between the site of disease onset and the spread of
phosphorylated, ubiquitinated, and N-terminally TDP-43 pathology in the spinal cord has been found.47
truncated (figure 3).1,44–46 ALS with dementia also involves accumulations of
TDP-43 in the extramotor neocortex and hippocampus.14,48
ALS-TDP The presence of pathological TDP-43 in a wide variety of
In most cases of sporadic ALS, the neuropathology is subcortical regions beyond the pyramidal system might
characterised by abnormal cytoplasmic accumulation of correlate with other clinical features, such as
TDP-43 in neurons and glia of the primary motor cortex, parkinsonism.42,49,50 Pathological TDP-43 seems to be a
brainstem motor nuclei, spinal cord, and the associated less consistent feature of other motor neuron disease
white matter tracts.14 Neuronal cytoplasmic inclusions phenotypes, such as primary lateral sclerosis and
in motor neurons vary in morphology and include progressive muscular atrophy,51 but further studies are
small granules, compact Lewy-body-like inclusions, and needed to confirm the degree of contribution.

www.thelancet.com/neurology Vol 9 October 2010 997


Review

ALS associated with mutations in TARDBP is reported with GRN mutations; subtype 2 generally presents with
to have TDP-43 pathology indistinguishable from that sporadic semantic dementia; subtype 3 is frequently
found in most sporadic cases.17,52–55 Several other causative associated with ALS with FTD, including familial cases
genes and genetically linked chromosomal loci have been linked to chromosome 9p; and subtype 4 is found only in
identified for familial ALS.56 TDP-43-positive ALS cases associated with VCP mutations.39,40,67,69,71–74
pathology in combination with FTLD-TDP has been
described in affected members of families with FTD and Other disorders with TDP-43 pathology
ALS linked to chromosome 9p.40 One case report of ALS Perry syndrome is a rare autosomal dominant disorder
associated with a mutation in ANG described TDP-43 characterised by parkinsonism, depression, weight loss,
pathology but with several atypical features.18 Mutations and hypoventilation and is associated with mutations in
in SOD1 and FUS are not associated with pathological exon 2 of DCTN1 encoding p150glued, the major subunit of
TDP-43.2,3,14,19,57–60 the dynactin protein complex.75 TDP-43 pathology is
largely restricted to the substantia nigra and other nuclei
FTLD-TDP of the rostral brainstem and the basal ganglia.76 Mutations
FTLD-TDP is the most frequent pathological FTLD in other regions of DCTN1 have previously been reported
subtype (~50% of cases) and can present with any of the in ALS with and without FTD,77–79 but it is unknown
major clinical phenotypes of the FTD syndrome, whether these disorders show TDP-43 pathology.
including behavioural variant FTD, semantic dementia, Concomitant TDP-43 pathology has been reported in
or progressive non-fluent aphasia.39,40 Extrapyramidal several other neurodegenerative disorders. This feature
movement disorders comprise a frequently seen minor is consistently seen in Guamanian ALS-parkinsonism-
feature, and FTLD-TDP rarely presents as Parkinson’s dementia complex,80,81 in some cases of hippocampal
disease or corticobasal syndrome.61–63 TDP-43 is also the sclerosis,82,83 and in 20–60% of cases of the most common
pathological substrate of most cases of dementia neurodegenerative disorders, including Alzheimer’s
combined with ALS.64,65 Although amnestic features are disease and Lewy body disorders where TDP-43 pathology
generally mild or emerge late in the course of FTD, some is mainly found in the mesial lobe.83–86
cases of FTLD-TDP present with an Alzheimer-like
phenotype.62 Genes and risk factors for TDP-43 proteinopathies
TDP-43-positive cytoplasmic inclusions and neurites TARDBP
in the frontotemporal neocortex and dentate granule In early 2008, mutations in TARDBP were identified in
cells of the hippocampus characterise FTLD-TDP a subset of patients with ALS.87–89 So far, 38 different
(figure 3).1,12,39,40 Lentiform neuronal intranuclear inclu- TARDBP mutations have been reported in
sions are seen more frequently in familial cases than in 79 genealogically unrelated families in the USA, Europe,
sporadic cases,39,66 and glial TDP-43 pathology is present Australia, and Asia. Mutations in this gene account for
in the subcortical white matter in all cases but to varying 4% of all familial and around 1·5% of sporadic ALS
degrees.41 TDP-43 pathology is also present in a wide cases (figure 2). Some evidence indicates that
range of subcortical neuroanatomical sites.48,67 Four frequencies of TARDBP mutations are higher among
different patterns of TDP-43 pathology are recognised in patients from southern Europe than among patients
FTLD-TDP, based on the anatomical distribution, from other regions.90
morphology, and relative proportion of distinct types of Most TARDBP mutations are missense changes in
inclusions (table).68–70 The relevance of this heterogeneity exon 6, encoding the Gly-rich region and C-terminus of
is supported by clinical and genetic correlations: subtype 1 TDP-43, and are suggested to interfere with normal
presents with either behavioural variant FTD or protein-protein interactions of TDP-43 (eg, by alteration
progressive non-fluent aphasia and represents all cases of phosphorylation or increase of aggregation tendency).

Type 1* Type 2* Type 3* Type 4


Neuronal cytoplasmic inclusions +++ + ++ +
Dystrophic neurites +++ +++ + +++
Neuronal intranuclear inclusions –/++ Generally absent Generally absent +++
Major clinical phenotypes bvFTD; PNFA SD bvFTD; FTD with ALS IBMPFD
Associated gene defects GRN mutations .. Linkage to chromosome 9p VCP mutations

Semiquantative grading is used: –, absent; +, few; ++, moderate; +++, numerous. ALS=amyotrophic lateral sclerosis. bvFTD=behavioural variant frontotemporal dementia.
FTD=frontotemporal dementia. FTLD=frontotemporal lobar degeneration. FTLD-U=FTLD with ubiquitinated inclusions. FTLD-TDP=TDP-43-positive FTLD. IBMPFD=inclusion
body myopathy associated with Paget disease of bone and frontotemporal dementia. PNFA=progressive non-fluent aphasia. SD=semantic dementia. *Numbering of types
according to MacKenzie et al.69 The same three patterns of FTLD-U pathology were independently described by Sampathu et al,68 but with a different numbering system.

Table: Neuropathological subtypes of FTLD-TDP

998 www.thelancet.com/neurology Vol 9 October 2010


Review

These effects might lead to impaired nuclear import, VCP


altered exon skipping, or transcriptional repression Mutations in the valosin containing protein gene (VCP)
activity.91 Two exceptions are the Asp169Gly mutation, cause a rare familial syndrome with TDP-43 pathology, in
which is located in exon 4, and the truncation mutation which the clinical features of inclusion body myopathy,
Tyr374X. These two mutations have, however, each been Paget’s disease of bone, and behavioural variant FTD
identified in only one patient with sporadic ALS and, show variable penetrance.117 A total of 14 different
therefore, their pathological roles remain unproven.88,92 missense mutations have been identified in 30 families.118
Despite extensive screening, TARDBP multiplications The link between VCP dysfunction and TDP-43
have not been identified.93–95 The clinical phenotype accumulation is unsolved, but transgenic mice expressing
associated with TARDBP mutations is classic adult-onset mutant forms of VCP develop pathology in muscle,
ALS with bulbar, limb, or respiratory symptoms, brain, and bone, recapitulating some features of the
although age of onset and duration vary.88,90,92,96 Three symptoms in humans.119
different TARDBP mutations have been reported in
patients with FTD: Gly295Ser in a patient with FTD Chromosome 9p
and ALS, Asn267Ser in a patient with behavioural An association with variation on the chromosomal locus
variant FTD, and Lys263Gln in a patient with 9p13.2–21.3 has been consistently identified in genome-
behavioural variant FTD with supranuclear gaze palsy wide linkage studies of autosomal dominant families in
and chorea. TARDBP mutations, therefore, might rarely which individual patients present with behavioural variant
cause FTD.15–17 FTD, ALS, or a combined phenotype.120–126 Despite extensive
genetic screening of the shared candidate region, the
GRN disease-associated gene has not yet been identified.
Mutations in the gene encoding the secreted growth Notably, a two-stage genome-wide association study that
factor progranulin (GRN) are the most frequent cause included data from nearly 20 000 patients with sporadic
of familial FTLD identified so far (up to 20%; 5–10% of ALS identified a significant genome-wide association with
all FTLD cases).97–99 68 different GRN mutations have UNC13A and a significant association in the replication
already been identified in more than 200 families. phase with a region on chromosome 9p, overlapping with For the FTD mutation database
Despite their dominant nature, GRN mutations all the locus associated with familial ALS with FTD.127 The see http://www.molgen.ua.ac.
be/FTDmutations/
cause disease through a haploinsufficiency/loss-of- association with the chromosome 9p locus has been
function mechanism.98,99 Although other types of confirmed in two independent genome-wide association
pathogenic mutations have been reported, most are studies.128,129 These findings strongly suggest that the
small out-of-frame insertions or deletions, splice-site, or disease gene is also a risk factor for sporadic ALS.
nonsense mutations that introduce a premature
termination codon and result in the degradation of the TMEM106B
mutant messenger RNA via nonsense-mediated Large cohorts of patients with FTD are rare, and the in-
decay.98–102 Of clinical relevance, concentrations of herent molecular heterogeneity had previously prohibited
progranulin in plasma, serum, and CSF are predictive genome-wide association studies. The identification of a
of GRN mutation status in mutation carriers with and pathological role for TDP-43, however, prompted a large
without symptoms.103–105 As a result, ELISA is expected to collaborative effort in which a homogeneous population
become an inexpensive alternative to GRN sequence of patients with pathologically confirmed FTLD-TDP
analysis, having the advantage of identifying patients was identified for such a study. Genome-wide associations
with rare mutations, such as single-exon deletions or were identified between FTLD-TDP and variants in
complete gene rearrangements, that would otherwise TMEM106B.130 The function of the TMEM106B protein is
be difficult to detect.105 currently unknown, but increased expression is suggested
FTLD patients with GRN mutations show notable to be a risk factor for developing FTLD-TDP.
variability in clinical phenotypes, including age of onset
and disease duration, even within individual families.106–109 Pathogenesis of TDP-43 proteinopathies
The most frequent presentations are behavioural variant Little is known about the role of TARDBP mutations or
FTD and progressive non-fluent aphasia. Memory TDP-43 pathology in neurodegeneration. The abnormal
impairment and extrapyramidal features are also intracellular redistribution of TDP-43 from the nucleus
frequently seen, but ALS is rare. Finally, common variants to the cytoplasm in inclusion-bearing neurons suggests a
in GRN have been reported as genetic risk factors for pathogenic mechanism associated with the loss of normal
TDP-43 proteinopathies.110–114 The exact mechanism by nuclear TDP-43 function.27,28,31 In support of this
which a loss of progranulin leads to TDP-43 pathology is hypothesis, knock down of TDP-43 in human cell lines
unknown. GRN knockdown in cell culture was reported induces morphological nuclear defects, altered neurite
to lead to increased cleavage of TDP-43 into potentially outgrowth, dysregulation of cell cycle, and increased cell
pathogenic C-terminal fragments,115 but this finding was death.131,132 Reduced TDP-43 expression in drosophila,
not confirmed in other studies.116 zebrafish, and mouse models resulted in motor deficits,

www.thelancet.com/neurology Vol 9 October 2010 999


Review

some with structural abnormalities of motor fully replicates the features of human disease, indicate
neurons.32,133,134 In one model, this phenotype could be that the pathogenic mechanism in TDP-43 proteinopathy
rescued by coexpression of wild-type TDP-43.133 is still unresolved. Given the multifaceted role of TDP-43,
Sequestration of TDP-43 in cellular inclusions could however, multiple disease mechanisms might be involved.
induce a toxic gain of function independently of the
basic biological role of this protein. A study in which FUS
TDP-43 was overexpressed in yeast suggested that only Normal function
aggregated TDP-43 was toxic.135 Several in-vitro studies FUS is a ubiquitously expressed 526 aminoacid protein
have shown that C-terminal fragments of TDP-43 are encoded by 15 exons that belongs to the FET/TET family
more likely than full-length TDP-43 to form insoluble of multifunctional DNA/RNA binding proteins (together
cytoplasmic aggregates that become ubiquitinated, with Ewing’s sarcoma protein and the TATA-binding
phosphorylated, and toxic to cells.136–139 Although one protein associated factor TAF15),148 and was originally
study suggested that these cytoplasmic aggregates bind discovered as a component of fusion oncogenes in
the endogenous full-length protein and deplete it from human cancers.149 It contains an N-terminal Gln-Gly-
the nucleus,137 another showed retention of normal Ser-Tyr-rich domain, a Gly-rich domain, an RNA
nuclear expression, suggesting a pure toxic effect.139 recognition motif, multiple Arg-Gly-Gly repeats, a zinc
Some in-vivo studies, in which wild-type human finger motif,149 and a highly conserved extreme
TDP-43 was overexpressed in drosophila,38,140 mice,141 C-terminus that encodes for a non-classic nuclear
and by viral expression in rats,142 have shown the localisation signal that is recognised by transportin
formation of TDP-43 immunoreactive neuronal cyto- (figure 2).150–152 FUS shows nuclear and cytoplasmic
plasmic inclusions with associated neurodegeneration expression,153 and continuously shuttles between the
and motor dysfunction. The findings, however, are nucleus and the cytoplasm.154 It is implicated in
inconsistent, because at least one study found no effect numerous cellular processes, including cell proliferation,
in transgenic rats overexpressing the wild-type human DNA repair, transcription regulation, and RNA and
protein.143 The two basic disease mechanisms are not microRNA processing,28,149,155 although its precise
mutually exclusive, because factors that affect the function is poorly characterised. This protein might be
normal intracellular trafficking of TDP-43 might involved in neuronal plasticity and maintenance of
predispose to the formation of abnormal aggregates and dendritic integrity by transporting messenger RNA to
the loss of nuclear localisation.26 dendritic spines for local translation.156,157 FUS knockout
The identification of TARDBP mutations in ALS has mice show variable phenotypes with perinatal lethality
provided additional clues to the possible pathogenic on an inbred genetic background158 but viability on an
mechanisms involved in TDP-43 proteinopathies. So outbred genetic background.159
far, no consequences on splicing activity, messenger
RNA processing, or protein binding have been Clinicopathological spectrum of FUS proteinopathies
described.30,137,144 Expression of mutant TDP-43 in FUS proteinopathies are characterised mainly by the
cultured cells is claimed to result in increased gen- presence of inclusions immunoreactive for FUS in
eration of C-terminal fragments, with even more neuronal and glial cells, but morphology and distribution
cytoplasmic aggregation and toxic effects than wild-type of inclusions vary between entities (figure 4). Re-
TDP-43.88,89,133,137,138,145,146 Results from transgenic animal distribution of FUS from the nucleus to the cytoplasm is
models overexpressing mutant forms of human TDP-43 seen to a lesser degree than for TDP-43, with at least
are somewhat inconsistent and difficult to interpret. In some of the nuclear staining being preserved in neurons
one study, transgenic rats overexpressing TDP-43 with with FUS inclusions.57,160 Despite increased FUS con-
the Met337Val mutation developed progressive weakness centrations in insoluble protein fractions, no obvious
and early death.143 Neurodegeneration was widespread disease-associated modifications of this protein, such as
but was most severe in the spinal cord. The human truncation, phosphorylation, or ubiquitination, have yet
TDP-43, however, was diffusely distributed in neuronal been identified.160
nuclei and cytoplasm, with cytoplasmic aggregates
being rare and present only in the cortex. Transgenic ALS-FUS
mice expressing the human Ala315Thr mutation have Mutations in the FUS gene were identified as the
also been reported to develop progressive motor primary cause of familial ALS linked to chromosome 16.2,3
abnormalities with widespread neurodegeneration.147 Patients with these mutations present with classic ALS
Although some neuronal populations were found to or might present with atypical phenotypes. Pathological
have cytoplasmic inclusions immunoreactive for data on FUS mutation carriers are available for only a
ubiquitin, these did not contain aggregated TDP-43 and few cases and only for single mutations.2,3,19,57,59,60,161 Initial
loss of nuclear TDP-43 was inconsistent. reports described motor neuron loss in the spinal cord
The inconsistencies among the various in-vitro and and to a lesser degree in the brainstem, with inconsistent
in-vivo models currently available, and the fact that none demyelination of corticospinal tracts and the presence

1000 www.thelancet.com/neurology Vol 9 October 2010


Review

A B C D E

G H I J

Figure 4: Pathological features in ALS-FUS and FTLD-FUS


FUS immunohistochemistry on paraffin-embedded tissue showing FUS-immunoreactive cytoplasmic inclusions in lower motor neurons of a familial ALS case with
the Arg521Cys FUS mutation, with (A) filamentous or (B) granular morphology. Nuclear FUS staining is present in the inclusion-bearing neuron (B). Three distinct
pathological entities of FTLD-FUS can be delineated. In atypical FTLD-U, FUS-immunoreactive cytoplasmic inclusions are detected in (C) the dentate granule cells and
(D) the frontal cortex, and (E, F) characteristic vermiform intranuclear inclusions are visible. Numerous FUS-immunoreactive cytoplasmic inclusions are detected with
variable morphology in the frontal cortex of (G) basophilic inclusion body disease and (H) neuronal intermediate filament inclusion disease. (I) Many inclusions in the
latter disorder are labelled only for FUS, and neurons with α-internexin-positive inclusions always show FUS pathology, with labelling of separate inclusion
components as demonstrated by double-label immunofluorescence for FUS (red) and α-internexin (green). (J) FUS-immunoreactive glial cytoplasmic inclusions are
present in ALS-FUS and FTLD-FUS. Scale bars: 30 μm (D, G, H), 20 μm (C), 15 μm (A, B), 8 μm (E, F, I, J). ALS=amyotrophic lateral sclerosis. FTLD=frontotemporal lobar
degeneration. FTLD-U=FTLD with ubiquitinated inclusions.

of FUS-positive inclusions3 or increased concentrations neurons.160,162–164 The most intriguing lesions in atypical
of cytoplasmic FUS2 in lower motor neurons. More FTLD-U are neuronal intranuclear inclusions, which
detailed pathological analysis of several cases with the appear as elongated straight, curved, or twisted
Arg521Cys mutation revealed more-widespread FUS (vermiform) filaments, or sometimes as ring-like
pathology, involving glial cells and regions outside the structures at the periphery of the nucleus and are most
pyramidal motor system, such as motor nuclei in the easily recognised in the hippocampus (figure 4).160
brainstem, striatum, thalamus, and substantia nigra. Basophilic inclusion body disease is a rare and clinically
The importance of extramotor pathology in ALS-FUS, heterogeneous disorder named for the presence of
as well as variability of pathological features among basophilic inclusions that are visible with haematoxylin
different FUS mutations, needs to be assessed in and eosin staining. It can present with behavioural
clinicopathological correlative studies. variant FTD,167–169 juvenile or adult-onset ALS,170–174 or a
combination of both.167,168,175,176 FUS immunohistochemistry
FTLD-FUS labels the basophilic inclusions and much more
FTLD-FUS consists of three distinct clinicopathological widespread, previously unrecognised neuronal and glial
entities (figure 1).13 Although all FTLD-U cases were pathology, predominantly in the frontal cortex, basal
thought to involve TDP-43, studies in large series of ganglia, brainstem, cerebellum, and spinal cord. Intra-
patients identified up to 20% of cases that were TDP-43 nuclear inclusions are usually absent.177
negative. This FTLD subtype was termed atypical Neuronal intermediate filament inclusion disease is
FTLD-U162,163 and is characterised by sporadic early-onset an uncommon neurodegenerative disorder that typically
behavioural variant FTD. Motor symptoms are not presents as early-onset sporadic behavioural variant
prominent, but if present they are typically limited to FTD that is associated with pyramidal or extrapyramidal
mild rigidity, intermittent hyperkinesias, or both.162–165 movement disorders, or both. This subtype was initially
In addition to frontotemporal atrophy severe caudate defined as a unique entity owing to the presence of
atrophy, and hippocampal sclerosis are characteristic neuronal inclusions immunoreactive for all class IV
features; caudate atrophy on neuroimaging has been intermediate filaments (light, medium, and heavy
proposed as a useful clinical predictor of this FTLD neurofilament subunits and α-internexin).178–180 The role
subtype.164,166 FUS pathology in atypical FTLD-U is most of intermediate filaments in the pathogenesis of
abundant in the frontotemporal neocortex and the neuronal intermediate filament inclusion disease, how-
hippocampus but is also present in the striatum, ever, is uncertain, because only a subset of inclusions
thalamus, and brainstem. Despite the absence of obvious are positive for these proteins. Antibodies against
clinical features of motor neuron disease, most cases FUS label many more inclusions than do those
reveal at least some FUS inclusions in lower motor against intermediate filaments.181 A large proportion of

www.thelancet.com/neurology Vol 9 October 2010 1001


Review

inclusions are only detected by FUS staining. By variants in controls, which indicates a benign role for
contrast, aggregates positive for intermediate filaments these polymorphisms.2,21,59,90
are always found in association with FUS pathology in Two FUS mutations have been reported that completely
the same cell (figure 4),181 thereby suggesting that FUS truncate the C-terminus: a splice-site mutation inducing
accumulation has a more central role in neuronal skipping of exon 14 (Gly466ValfsX14) and a nonsense
intermediate filament inclusion disease than does mutation in exon 14 (Arg495X).187,192 The mutation
accumulation of intermediate filaments, which might Gly466ValfsX14 has occurred de novo, leading to sporadic
be a secondary phenomenon, possibly caused by altered disease.187 Because relatives of sporadic FUS mutation
messenger RNA processing or trafficking. carriers have been unavailable in most studies, the true
frequency of de-novo FUS mutations remains to be
Other disorders with FUS pathology determined.
FUS immunoreactivity has been described in the Patients with FUS mutations generally present with
intranuclear inclusions in a wide spectrum of classic ALS, although some develop very little or no upper
polyglutamine diseases, such as Huntington’s disease motor neuron signs. Some studies have associated
and spinocerebellar ataxias.182,183 The functional relevance specific phenotypes with certain FUS mutations
of FUS accumulation in these disorders, however, (eg, symmetric weakness of proximal muscle groups
remains to be determined. with only late involvement of distal muscles and
minimum evidence of upper motor neuron involvement
Spectrum of FUS gene mutations for the common Arg521Cys mutation).19,20,57,189,193 The
FUS was deemed to be a candidate gene for familial ALS identification of additional families carrying the same
linked to chromosome 16 on the basis of its homologous mutation should provide more insight into the
domain structure to TDP-43.2,3 In only 2 years, 35 different correlations between genotypes and phenotypes. Onset
pathogenic FUS mutations have been identified that age varies strikingly from early 20s to late 70s, even
account for about 4% of familial ALS cases, but less than within families.57 For some mutations non-penetrance
1% of sporadic ALS cases (figure 2).2,3,19,20,57,59,60,161,184–192 has been described with mutation carriers surviving to
Mutation carriers have been reported in the USA, Europe, age 80 years without signs of ALS,2,59,185 a finding that
Australia, Africa, and Asia. The mutation frequency in should be taken into consideration when providing
familial ALS cohorts has varied from 0 to 10%, and might genetic counselling for FUS mutation carriers.
depend on the geographical origin of patients, the Reports of FTD associated with FUS mutations are
numbers screened, or the fact that only a subset of exons rare. Two patients had ALS with FTD features.19,20
was analysed.161,185–187,191,192 Mutations in FUS are mostly Although one patient with the Met254Val mutation
missense changes that cluster in two regions: most are presented with behavioural variant FTD, both parents
located in exons 13–15, which encode the Arg-Gly-Gly-rich were still alive and asymptomatic, and no autopsy data
region and the nuclear localisation signal of FUS, and were available to confirm FUS pathology.21 Additional
about a third of the mutations are located in exons 3, 5, studies will, therefore, be needed to determine whether
and 6, which encode the Gln-Gly-Ser-Tyr-rich and Gly-rich FUS is a true cause of FTD.
regions. Mutations affecting the C-terminus are clearly In sharp contrast to ALS-FUS, where genetic
pathogenic, with several of them leading to disruption of sequencing has shown FUS mutations in almost all
the nuclear localisation signal motif, which impairs cases, extensive analysis of genomic DNA, complementary
transportin-mediated nuclear import.152 C-terminal DNA, and messenger RNA in patients with FTLD-FUS
mutations have so far been found in 54 patients with has thus far excluded FUS mutations.160,165,181 Although
familial ALS, but in only four patients with sporadic this finding supports the sporadic nature of FTLD-FUS
ALS.2,3,19,20,57,59,60,161,184–192 By contrast, seven of 11 patients with in most patients, the presence of recessive or de-novo
mutations affecting exons 3, 5, or 6 had sporadic disease mutations in FUS or other genes cannot be excluded.
and no segregation with disease was seen for the other
four familial cases.2,20,57,189,191 Although several of these Pathogenesis of FUS proteinopathies
mutations in exons 3, 5, and 6 affect evolutionary Understanding of the underlying pathomechanisms
conserved aminoacids and are predicted to be harmful on leading to FUS accumulation and FUS-mediated
the basis of findings of in-silico analysis, no functional neurodegeneration is still limited. As for TDP-43
consequences have yet been shown.152 Moreover, no proteinopathies, a toxic gain-of-function mechanism, a
autopsy data showing FUS pathology are yet available for loss-of-function mechanism by depletion of physiological
these mutations, which leaves open the possibility that FUS and maybe co-sequestration of other vital factors, or
they might be risk factors rather than true causal both is plausible. So far, no in-vivo models for FUS
mutations. In addition to missense mutations, several proteinopathies have been reported. Expression of
in-frame insertions and deletions affect the Gly stretches C-terminal FUS missense and truncation mutations
encoded by exons 5 and 6. Although initially suggested to in vitro have revealed variable increases in cytoplasmic
cause disease, subsequent studies identified similar concentrations of FUS2,3,187 that are compatible with

1002 www.thelancet.com/neurology Vol 9 October 2010


Review

Contributors
Search strategy and selection criteria All authors contributed equally to the conception, design, literature
search, and writing of this Review.
We searched PubMed to identify full-text articles published
Conflicts of interest
up to June, 2010, with the terms “frontotemporal dementia”, We declare that we have no conflicts of interest.
“amyotrophic lateral sclerosis”, “TDP-43”, “TARDBP”, “FUS”,
Acknowledgments
and “TLS”. Articles were also identified through searches of We thank Richard Crook, Department of Neuroscience, Mayo Clinic
the authors’ own article collections. Only articles published in College of Medicine, Jacksonville, FL, USA, for help on figure 2.
English were reviewed. Work relevant to this Review was funded by grants from the National
Institutes of Health/National Institute on Aging (AG26251-03A1 to
RR), the ALS Association (RR), the German Federal Ministry of
Education and Research (MN, grant number 01GI0704), the Synapsis
disruption or deletion of the nuclear localisation signal Foundation (MN), Canadian Institutes of Health Research (IRAM,
and impaired transportin-mediated nuclear import,152 grant number 74580), and the Pacific Alzheimer Research
thereby implicating altered nuclear import as a key event Foundation (IRAM).
in disease pathogenesis. Notably, the two reported References
truncation mutations led to very aggressive and early- 1 Neumann M, Sampathu DM, Kwong LK, et al. Ubiquitinated
TDP-43 in frontotemporal lobar degeneration and amyotrophic
onset forms of ALS,187,192 which might reflect their severe lateral sclerosis. Science 2006; 314: 130–33.
impairment of nuclear import. Further studies are needed 2 Kwiatkowski TJ Jr, Bosco DA, Leclerc AL, et al. Mutations in the
to address whether and how additional domains and post- FUS/TLS gene on chromosome 16 cause familial amyotrophic
lateral sclerosis. Science 2009; 323: 1205–08.
translational modifications modulate nuclear-cytoplasmic
3 Vance C, Rogelj B, Hortobagyi T, et al. Mutations in FUS, an RNA
shuttling of FUS. Modifications of FUS under specific processing protein, cause familial amyotrophic lateral sclerosis
conditions include phosphorylation194,195 and arginine type 6. Science 2009; 323: 1208–11.
methylation.196,197 Although the associated functional 4 Mitchell JD, Borasio GD. Amyotrophic lateral sclerosis. Lancet 2007;
369: 2031–41.
consequences have not yet been elucidated for FUS, 5 Rosen DR, Siddique T, Patterson D, et al. Mutations in Cu/Zn
similar modifications of other RNA binding proteins superoxide dismutase gene are associated with familial amyotrophic
affect protein-protein or protein-RNA interactions that lateral sclerosis. Nature 1993; 362: 59–62.
6 Boillee S, Vande Velde C, Cleveland DW. ALS: a disease of motor
are crucial for nuclear-cytoplasmic shuttling.198,199 Although neurons and their nonneuronal neighbors. Neuron 2006;
the ways in which impaired nuclear import of FUS 52: 39–59.
mediates neurodegeneration remain unclear, recruitment 7 Bruijn LI, Houseweart MK, Kato S, et al. Aggregation and motor
of FUS into stress granules upon interference with neuron toxicity of an ALS-linked SOD1 mutant independent from
wild-type SOD1. Science 1998; 281: 1851–54.
nuclear import in vitro implicates formation of stress 8 McKhann GM, Albert MS, Grossman M, Miller B, Dickson D,
granules in disease pathogenesis.152 This hypothesis is Trojanowski JQ. Clinical and pathological diagnosis of
supported by the presence of stress granule markers in frontotemporal dementia: report of the Work Group on
Frontotemporal Dementia and Pick’s Disease. Arch Neurol 2001;
human FUS inclusions.152,173 58: 1803–09.
9 Lomen-Hoerth C, Anderson T, Miller B. The overlap of amyotrophic
Conclusions and future directions lateral sclerosis and frontotemporal dementia. Neurology 2002;
59: 1077–1079.
The seminal discoveries of two novel key players, TDP-43 10 Murphy JM, Henry RG, Langmore S, Kramer JH, Miller BL,
and FUS, have led to a notable shift in the understanding Lomen-Hoerth C. Continuum of frontal lobe impairment in
of pathomechanisms in ALS and FTD, and have opened amyotrophic lateral sclerosis. Arch Neurol 2007; 64: 530–34.
new avenues for research. The structural and functional 11 Mackenzie IR, Feldman HH. Ubiquitin immunohistochemistry
suggests classic motor neuron disease, motor neuron disease with
similarities of these proteins implicate alterations in dementia, and frontotemporal dementia of the motor neuron
processing of RNA and microRNA as key events in disease type represent a clinicopathologic spectrum.
J Neuropathol Exp Neurol 2005; 64: 730–39.
disease development, although the underlying functional
12 Arai T, Hasegawa M, Akiyama H, et al. TDP-43 is a component of
mechanisms remain unresolved. So far, all studies except ubiquitin-positive tau-negative inclusions in frontotemporal lobar
one200 have shown TDP-43 and FUS pathology to be degeneration and amyotrophic lateral sclerosis.
Biochem Biophys Res Commun 2006; 351: 602–11.
mutually exclusive, thereby implying independent disease
13 Mackenzie IR, Neumann M, Bigio EH, et al. Nomenclature and
pathways.3,59,160,164 However, the possibility of interactions nosology for neuropathologic subtypes of frontotemporal lobar
between TDP-43 and FUS and their associations with the degeneration: an update. Acta Neuropathol 2010; 119: 1–4.
protein optineurin, which has been related to familial 14 Mackenzie IR, Bigio EH, Ince PG, et al. Pathological TDP-43
distinguishes sporadic amyotrophic lateral sclerosis from
ALS,201 needs to be addressed in more detail. Future efforts amyotrophic lateral sclerosis with SOD1 mutations. Ann Neurol
should focus on the generation and establishment of valid 2007; 61: 427–34.
in-vivo models, the identification of disease-relevant post- 15 Borroni B, Bonvicini C, Alberici A, et al. Mutation within TARDBP
leads to frontotemporal dementia without motor neuron disease.
translational modifications, and brain-specific messenger Hum Mutat 2009; 30: E974–83.
RNA and microRNA targets of both proteins. From the 16 Benajiba L, Le Ber I, Camuzat A, et al. TARDBP mutations in
clinical perspective, TDP-43 and FUS might be promising motoneuron disease with frontotemporal lobar degeneration.
Ann Neurol 2009; 65: 470–73.
candidates for the development of novel biomarker assays
17 Kovacs GG, Murrell JR, Horvath S, et al. TARDBP variation
for the diagnosis202,203 and progression of disease, and for associated with frontotemporal dementia, supranuclear gaze palsy,
the development of targeted therapies. and chorea. Mov Disord 2009; 24: 1843–47.

www.thelancet.com/neurology Vol 9 October 2010 1003


Review

18 Seilhean D, Cazeneuve C, Thuries V, et al. Accumulation of TDP-43 41 Neumann M, Kwong LK, Truax AC, et al. TDP-43-positive white
and alpha-actin in an amyotrophic lateral sclerosis patient with the matter pathology in frontotemporal lobar degeneration with ubiquitin-
K17I ANG mutation. Acta Neuropathol 2009; 118: 561–73. positive inclusions. J Neuropathol Exp Neurol 2007; 66: 177–83.
19 Blair IP, Williams KL, Warraich ST, et al. FUS mutations in 42 Brandmeir NJ, Geser F, Kwong LK, et al. Severe subcortical TDP-43
amyotrophic lateral sclerosis: clinical, pathological, neurophysiological pathology in sporadic frontotemporal lobar degeneration with
and genetic analysis. J Neurol Neurosurg Psychiatry 2009; 81: 639–41. motor neuron disease. Acta Neuropathol 2008; 115: 123–31.
20 Ticozzi N, Silani V, LeClerc AL, et al. Analysis of FUS gene 43 Hatanpaa KJ, Bigio EH, Cairns NJ, et al. TAR DNA-binding
mutation in familial amyotrophic lateral sclerosis within an Italian protein 43 immunohistochemistry reveals extensive neuritic
cohort. Neurology 2009; 73: 1180–85. pathology in FTLD-U: a Midwest-Southwest Consortium for FTLD
21 Van Langenhove T, van der Zee J, Sleegers K, et al. Genetic Study. J Neuropathol Exp Neurol 2008; 67: 271–79.
contribution of FUS to frontotemporal lobar degeneration. 44 Hasegawa M, Arai T, Nonaka T, et al. Phosphorylated TDP-43 in
Neurology 2010; 74: 366–71. frontotemporal lobar degeneration and amyotrophic lateral
22 Buratti E, Dork T, Zuccato E, Pagani F, Romano M, Baralle FE. sclerosis. Ann Neurol 2008; 64: 60–70.
Nuclear factor TDP-43 and SR proteins promote in vitro and in vivo 45 Neumann M, Kwong LK, Lee EB, et al. Phosphorylation of S409/410
CFTR exon 9 skipping. EMBO J 2001; 20: 1774–84. of TDP-43 is a consistent feature in all sporadic and familial forms
23 Wang HY, Wang IF, Bose J, Shen CK. Structural diversity and of TDP-43 proteinopathies. Acta Neuropathol 2009; 117: 137–49.
functional implications of the eukaryotic TDP gene family. 46 Igaz LM, Kwong LK, Xu Y, et al. Enrichment of C-terminal
Genomics 2004; 83: 130–39. fragments in TAR DNA-binding protein-43 cytoplasmic inclusions
24 Buratti E, Brindisi A, Giombi M, Tisminetzky S, Ayala YM, in brain but not in spinal cord of frontotemporal lobar degeneration
Baralle FE. TDP-43 binds heterogeneous nuclear ribonucleoprotein and amyotrophic lateral sclerosis. Am J Pathol 2008; 173: 182–94.
A/B through its C-terminal tail: an important region for the 47 Bodansky A, Kim JM, Tempest L, Velagapudi A, Libby R, Ravits J.
inhibition of cystic fibrosis transmembrane conductance regulator TDP-43 and ubiquitinated cytoplasmic aggregates in sporadic ALS
exon 9 splicing. J Biol Chem 2005; 280: 37572–84. are low frequency and widely distributed in the lower motor neuron
25 Ayala YM, Zago P, D’Ambrogio A, et al. Structural determinants of columns independent of disease spread. Amyotroph Lateral Scler
the cellular localization and shuttling of TDP-43. J Cell Sci 2008; 2010; 11: 321–27.
121: 3778–85. 48 Geser F, Martinez-Lage M, Robinson J, et al. Clinical and
26 Winton MJ, Igaz LM, Wong MM, Kwong LK, Trojanowski JQ, pathological continuum of multisystem TDP-43 proteinopathies.
Lee VM. Disturbance of nuclear and cytoplasmic TAR DNA-binding Arch Neurol 2009; 66: 180–89.
protein (TDP-43) induces disease-like redistribution, sequestration, 49 Geser F, Brandmeir NJ, Kwong LK, et al. Evidence of multisystem
and aggregate formation. J Biol Chem 2008; 283: 13302–09. disorder in whole-brain map of pathological TDP-43 in amyotrophic
27 Buratti E, Baralle FE. Multiple roles of TDP-43 in gene expression, lateral sclerosis. Arch Neurol 2008; 65: 636–41.
splicing regulation, and human disease. Front Biosci 2008; 13: 867–78. 50 Zhang H, Tan CF, Mori F, et al. TDP-43-immunoreactive neuronal
28 Lagier-Tourenne C, Polymenidou M, Cleveland DW. TDP-43 and and glial inclusions in the neostriatum in amyotrophic lateral
FUS/TLS: emerging roles in RNA processing and sclerosis with and without dementia. Acta Neuropathol 2008;
neurodegeneration. Hum Mol Genet 2010; 19: R46–64. 115: 115–22.
29 Buratti E, De Conti L, Stuani C, Romano M, Baralle M, Baralle F. 51 Dickson DW, Josephs KA, Amador-Ortiz C. TDP-43 in differential
Nuclear factor TDP-43 can affect selected microRNA levels. FEBS J diagnosis of motor neuron disorders. Acta Neuropathol (Berl) 2007;
2010; 277: 2268–81. 114: 71–79.
30 Fiesel FC, Voigt A, Weber SS, et al. Knockdown of transactive 52 Pamphlett R, Luquin N, McLean C, Jew SK, Adams L. TDP-43
response DNA-binding protein (TDP-43) downregulates histone neuropathology is similar in sporadic amyotrophic lateral sclerosis
deacetylase 6. EMBO J 2010; 29: 209–21. with or without TDP-43 mutations. Neuropathol Appl Neurobiol
31 Strong MJ, Volkening K, Hammond R, et al. TDP43 is a human low 2009; 35: 222–25.
molecular weight neurofilament (hNFL) mRNA-binding protein. 53 Yokoseki A, Shiga A, Tan CF, et al. TDP-43 mutation in familial
Mol Cell Neurosci 2007; 35: 320–27. amyotrophic lateral sclerosis. Ann Neurol 2008; 63: 538–42.
32 Kraemer BC, Schuck T, Wheeler JM, et al. Loss of murine TDP-43 54 Van Deerlin VM, Leverenz JB, Bekris LM, et al. TARDBP mutations
disrupts motor function and plays an essential role in in amyotrophic lateral sclerosis with TDP-43 neuropathology: a
embryogenesis. Acta Neuropathol 2010; 119: 409–19. genetic and histopathological analysis. Lancet Neurol 2008; 7: 409–16.
33 Wu LS, Cheng WC, Hou SC, Yan YT, Jiang ST, Shen CK. TDP-43, 55 Tamaoka A, Arai M, Itokawa M, et al. TDP-43 M337V mutation in
a neuro-pathosignature factor, is essential for early mouse familial amyotrophic lateral sclerosis in Japan. Intern Med 2010;
embryogenesis. Genesis 2010; 48: 56–62. 49: 331–34.
34 Sephton CF, Good SK, Atkin S, et al. TDP-43 is a developmentally 56 Valdmanis PN, Rouleau GA. Genetics of familial amyotrophic
regulated protein essential for early embryonic development. lateral sclerosis. Neurology 2008; 70: 144–52.
J Biol Chem 2010; 285: 6826–34. 57 Rademakers R, Stewart H, DeJesus-Hernandez M, et al. FUS gene
35 Moisse K, Mepham J, Volkening K, Welch I, Hill T, Strong MJ. mutations in familial and sporadic amyotrophic lateral sclerosis.
Cytosolic TDP-43 expression following axotomy is associated with Muscle Nerve 2010; 42: 170–76.
caspase 3 activation in NFL-/- mice: support for a role for TDP-43 in 58 Tan CF, Eguchi H, Tagawa A, et al. TDP-43 immunoreactivity in
the physiological response to neuronal injury. Brain Res 2009; neuronal inclusions in familial amyotrophic lateral sclerosis with or
1296: 176–86. without SOD1 gene mutation. Acta Neuropathol (Berl) 2007;
36 Sato T, Takeuchi S, Saito A, et al. Axonal ligation induces transient 113: 535–42.
redistribution of TDP-43 in brainstem motor neurons. Neuroscience 59 Hewitt C, Kirby J, Highley JR, et al. Novel FUS/TLS mutations and
2009; 164: 1565–78. pathology in familial and sporadic amyotrophic lateral sclerosis.
37 Wang IF, Wu LS, Chang HY, Shen CK. TDP-43, the signature Arch Neurol 2010; 67: 455–61.
protein of FTLD-U, is a neuronal activity-responsive factor. 60 Tateishi T, Hokonohara T, Yamasaki R, et al. Multiple system
J Neurochem 2008; 105: 797–806. degeneration with basophilic inclusions in Japanese ALS patients
38 Lu Y, Ferris J, Gao FB. Frontotemporal dementia and amyotrophic with FUS mutation. Acta Neuropathol 2010; 119: 355–64.
lateral sclerosis-associated disease protein TDP-43 promotes 61 Spina S, Murrell JR, Huey ED, et al. Clinicopathologic features of
dendritic branching. Mol Brain 2009; 2: 30. frontotemporal dementia with progranulin sequence variation.
39 Davidson Y, Kelley T, Mackenzie IR, et al. Ubiquitinated Neurology 2007; 68: 820–27.
pathological lesions in frontotemporal lobar degeneration contain 62 Gass J, Cannon A, Mackenzie IR, et al. Mutations in progranulin
the TAR DNA-binding protein, TDP-43. Acta Neuropathol (Berl) are a major cause of ubiquitin-positive frontotemporal lobar
2007; 113: 521–33. degeneration. Hum Mol Genet 2006; 15: 2988–3001.
40 Cairns NJ, Neumann M, Bigio EH, et al. TDP-43 in familial and 63 Masellis M, Momeni P, Meschino W, et al. Novel splicing mutation
sporadic frontotemporal lobar degeneration with ubiquitin in the progranulin gene causing familial corticobasal syndrome.
inclusions. Am J Pathol 2007; 171: 227–40. Brain 2006; 129: 3115–23.

1004 www.thelancet.com/neurology Vol 9 October 2010


Review

64 Josephs KA, Parisi JE, Knopman DS, Boeve BF, Petersen RC, 88 Kabashi E, Valdmanis PN, Dion P, et al. TARDBP mutations in
Dickson DW. Clinically undetected motor neuron disease in individuals with sporadic and familial amyotrophic lateral sclerosis.
pathologically proven frontotemporal lobar degeneration with Nat Genet 2008; 40: 572–74.
motor neuron disease. Arch Neurol 2006; 63: 506–12. 89 Sreedharan J, Blair IP, Tripathi VB, et al. TDP-43 mutations in
65 Snowden J, Neary D, Mann D. Frontotemporal lobar degeneration: familial and sporadic amyotrophic lateral sclerosis. Science 2008;
clinical and pathological relationships. Acta Neuropathol 2007; 319: 1668–72.
114: 31–38. 90 Corrado L, Ratti A, Gellera C, et al. High frequency of TARDBP
66 Mackenzie IR, Feldman H. Neuronal intranuclear inclusions gene mutations in Italian patients with amyotrophic lateral
distinguish familial FTD-MND type from sporadic cases. sclerosis. Hum Mutat 2009; 30: 688–94.
Acta Neuropathol(Berl) 2003; 105: 543–48. 91 Pesiridis GS, Lee VM, Trojanowski JQ. Mutations in TDP-43 link
67 Josephs KA, Stroh A, Dugger B, Dickson DW. Evaluation of glycine-rich domain functions to amyotrophic lateral sclerosis.
subcortical pathology and clinical correlations in FTLD-U subtypes. Hum Mol Genet 2009; 18: R156–62.
Acta Neuropathol 2009; 118: 349–58. 92 Daoud H, Valdmanis PN, Kabashi E, et al. Contribution of TARDBP
68 Sampathu DM, Neumann M, Kwong LK, et al. Pathological mutations to sporadic amyotrophic lateral sclerosis. J Med Genet
heterogeneity of frontotemporal lobar degeneration with ubiquitin- 2009; 46: 112–14.
positive inclusions delineated by ubiquitin immunohistochemistry 93 Rutherford NJ, Zhang YJ, Baker M, et al. Novel mutations in
and novel monoclonal antibodies. Am J Pathol 2006; 169: 1343–52. TARDBP (TDP-43) in patients with familial amyotrophic lateral
69 Mackenzie IR, Baborie A, Pickering-Brown S, et al. Heterogeneity sclerosis. PLoS Genet 2008; 4: e1000193.
of ubiquitin pathology in frontotemporal lobar degeneration: 94 Baumer D, Parkinson N, Talbot K. TARDBP in amyotrophic lateral
classification and relation to clinical phenotype. sclerosis: identification of a novel variant but absence of copy
Acta Neuropathol (Berl) 2006; 112: 539–49. number variation. J Neurol Neurosurg Psychiatry 2009; 80: 1283–85.
70 Cairns NJ, Bigio EH, Mackenzie IR, et al. Neuropathologic 95 Gijselinck I, Sleegers K, Engelborghs S, et al. Neuronal inclusion
diagnostic and nosologic criteria for frontotemporal lobar protein TDP-43 has no primary genetic role in FTD and ALS.
degeneration: consensus of the Consortium for Frontotemporal Neurobiol Aging 2009; 30: 1329–31.
Lobar Degeneration. Acta Neuropathol (Berl) 2007; 114: 5–22. 96 Kirby J, Goodall EF, Smith W, et al. Broad clinical phenotypes
71 Mackenzie IR, Baker M, Pickering-Brown S, et al. The associated with TAR-DNA binding protein (TARDBP) mutations in
neuropathology of frontotemporal lobar degeneration caused by amyotrophic lateral sclerosis. Neurogenetics 2010; 11: 217–25.
mutations in the progranulin gene. Brain 2006; 129: 3081–90. 97 Rademakers R, Hutton M. The genetics of frontotemporal lobar
72 Forman MS, Mackenzie IR, Cairns NJ, et al. Novel ubiquitin degeneration. Curr Neurol Neurosci Rep 2007; 7: 434–42.
neuropathology in frontotemporal dementia with valosin-containing 98 Cruts M, Gijselinck I, van der Zee J, et al. Null mutations in
protein gene mutations. J Neuropathol Exp Neurol 2006; 65: 571–81. progranulin cause ubiquitin-positive frontotemporal dementia
73 Neumann M, Mackenzie IR, Cairns NJ, et al. TDP-43 in the linked to chromosome 17q21. Nature 2006; 442: 920–24.
ubiquitin pathology of frontotemporal dementia with VCP gene 99 Baker M, Mackenzie IR, Pickering-Brown SM, et al. Mutations in
mutations. J Neuropathol Exp Neurol 2007; 66: 152–57. progranulin cause tau-negative frontotemporal dementia linked to
74 Josephs KA, Ahmed Z, Katsuse O, et al. Neuropathologic features of chromosome 17. Nature 2006; 442: 916–19.
frontotemporal lobar degeneration with ubiquitin-positive 100 Rademakers R, Rovelet-Lecrux A. Recent insights into the
inclusions with progranulin gene (PGRN) mutations. molecular genetics of dementia. Trends Neurosci 2009; 32: 451–61.
J Neuropathol Exp Neurol 2007; 66: 142–51.
101 Mukherjee O, Pastor P, Cairns NJ, et al. HDDD2 is a familial
75 Farrer MJ, Hulihan MM, Kachergus JM, et al. DCTN1 mutations in frontotemporal lobar degeneration with ubiquitin-positive, tau-
Perry syndrome. Nat Genet 2009; 41: 163–65. negative inclusions caused by a missense mutation in the signal
76 Wider C, Dickson DW, Stoessl AJ, et al. Pallidonigral TDP-43 peptide of progranulin. Ann Neurol 2006; 60: 314–22.
pathology in Perry syndrome. Parkinsonism Relat Disord 2009; 102 Gijselinck I, van der Zee J, Engelborghs S, et al. Progranulin locus
15: 281–86. deletion in frontotemporal dementia. Hum Mutat 2008; 29: 53–58.
77 Puls I, Jonnakuty C, LaMonte BH, et al. Mutant dynactin in motor 103 Sleegers K, Brouwers N, Van Damme P, et al. Serum biomarker for
neuron disease. Nat Genet 2003; 33: 455–56. progranulin-associated frontotemporal lobar degeneration.
78 Munch C, Sedlmeier R, Meyer T, et al. Point mutations of the p150 Ann Neurol 2009; 65: 603–09.
subunit of dynactin (DCTN1) gene in ALS. Neurology 2004; 63: 724–26. 104 Ghidoni R, Benussi L, Glionna M, Franzoni M, Binetti G. Low
79 Munch C, Rosenbohm A, Sperfeld AD, et al. Heterozygous R1101K plasma progranulin levels predict progranulin mutations in
mutation of the DCTN1 gene in a family with ALS and FTD. frontotemporal lobar degeneration. Neurology 2008; 71: 1235–39.
Ann Neurol 2005; 58: 777–80. 105 Finch N, Baker M, Crook R, et al. Plasma progranulin levels
80 Geser F, Winton MJ, Kwong LK, et al. Pathological TDP-43 in predict progranulin mutation status in frontotemporal dementia
parkinsonism-dementia complex and amyotrophic lateral sclerosis patients and asymptomatic family members. Brain 2009;
of Guam. Acta Neuropathol 2008; 115: 133–45. 132: 583–91.
81 Hasegawa M, Arai T, Akiyama H, et al. TDP-43 is deposited in the 106 Rademakers R, Baker M, Gass J, et al. Phenotypic variability
Guam parkinsonism-dementia complex brains. Brain 2007; associated with progranulin haploinsufficiency in patients with the
130: 1386–94. common 1477C→T (Arg493X) mutation: an international initiative.
82 Probst A, Taylor KI, Tolnay M. Hippocampal sclerosis dementia: Lancet Neurol 2007; 6: 857–68.
a reappraisal. Acta Neuropathol (Berl) 2007; 114: 335–45. 107 Le Ber I, Camuzat A, Hannequin D, et al. Phenotype variability in
83 Amador-Ortiz C, Lin WL, Ahmed Z, et al. TDP-43 immunoreactivity progranulin mutation carriers: a clinical, neuropsychological,
in hippocampal sclerosis and Alzheimer’s disease. Ann Neurol 2007; imaging and genetic study. Brain 2008; 131: 732–46.
61: 435–45. 108 Benussi L, Ghidoni R, Pegoiani E, Moretti DV, Zanetti O, Binetti G.
84 Uryu K, Nakashima-Yasuda H, Forman MS, et al. Concomitant Progranulin Leu271LeufsX10 is one of the most common FTLD
TAR-DNA-binding protein 43 pathology is present in Alzheimer and CBS associated mutations worldwide. Neurobiol Dis 2009;
disease and corticobasal degeneration but not in other tauopathies. 33: 379–85.
J Neuropathol Exp Neurol 2008; 67: 555–64. 109 Kelley BJ, Haidar W, Boeve BF, et al. Prominent phenotypic
85 Arai T, Mackenzie IR, Hasegawa M, et al. Phosphorylated TDP-43 variability associated with mutations in progranulin.
in Alzheimer’s disease and dementia with Lewy bodies. Neurobiol Aging 2009; 30: 739–51.
Acta Neuropathol 2009; 117: 125–36. 110 Rademakers R, Eriksen JL, Baker M, et al. Common variation in the
86 Higashi S, Iseki E, Yamamoto R, et al. Concurrence of TDP-43, tau miR-659 binding-site of GRN is a major risk factor for TDP43-positive
and alpha-synuclein pathology in brains of Alzheimer’s disease and frontotemporal dementia. Hum Mol Genet 2008; 17: 3631–42.
dementia with Lewy bodies. Brain Res 2007; 1184: 284–94. 111 Dickson DW, Baker M, Rademakers R. Common variant in GRN is
87 Gitcho MA, Baloh RH, Chakraverty S, et al. TDP-43 A315T mutation a genetic risk factor for hippocampal sclerosis in the elderly.
in familial motor neuron disease. Ann Neurol 2008; 63: 535–38. Neurodegener Dis 2010; 7: 170–74.

www.thelancet.com/neurology Vol 9 October 2010 1005


Review

112 Sleegers K, Brouwers N, Maurer-Stroh S, et al. Progranulin genetic 134 Feiguin F, Godena VK, Romano G, D’Ambrogio A, Klima R,
variability contributes to amyotrophic lateral sclerosis. Neurology Baralle FE. Depletion of TDP-43 affects Drosophila motoneurons
2008; 71: 253–59. terminal synapsis and locomotive behavior. FEBS Lett 2009;
113 Galimberti D, Fenoglio C, Cortini F, et al. GRN variability 583: 1586–92.
contributes to sporadic frontotemporal lobar degeneration. 135 Johnson BS, McCaffery JM, Lindquist S, Gitler AD. A yeast TDP-43
J Alzheimers Dis 2010; 19: 171–77. proteinopathy model: exploring the molecular determinants of
114 Brouwers N, Sleegers K, Engelborghs S, et al. Genetic variability in TDP-43 aggregation and cellular toxicity. Proc Natl Acad Sci USA
progranulin contributes to risk for clinically diagnosed Alzheimer 2008; 105: 6439–44.
disease. Neurology 2008; 71: 656–64. 136 Igaz LM, Kwong LK, Chen-Plotkin A, et al. Expression of TDP-43
115 Zhang YJ, Xu YF, Dickey CA, et al. Progranulin mediates caspase- C-terminal fragments in vitro recapitulates pathological features of
dependent cleavage of TAR DNA binding protein-43. J Neurosci TDP-43 proteinopathies. J Biol Chem 2009; 284: 8516–24.
2007; 27: 10530–34. 137 Nonaka T, Kametani F, Arai T, Akiyama H, Hasegawa M.
116 Dormann D, Capell A, Carlson AM, et al. Proteolytic processing of Truncation and pathogenic mutations facilitate the formation of
TAR DNA binding protein-43 by caspases produces C-terminal intracellular aggregates of TDP-43. Hum Mol Genet 2009;
fragments with disease defining properties independent of 18: 3353–64.
progranulin. J Neurochem 2009; 110: 1082–94. 138 Arai T, Hasegawa M, Nonoka T, et al. Phosphorylated and cleaved
117 Watts GD, Wymer J, Kovach MJ, et al. Inclusion body myopathy TDP-43 in ALS, FTLD and other neurodegenerative disorders and
associated with Paget disease of bone and frontotemporal dementia in cellular models of TDP-43 proteinopathy. Neuropathology 2010;
is caused by mutant valosin-containing protein. Nat Genet 2004; 30: 170–81.
36: 377–81. 139 Zhang YJ, Xu YF, Cook C, et al. Aberrant cleavage of TDP-43
118 Kimonis VE, Fulchiero E, Vesa J, Watts G. VCP disease associated enhances aggregation and cellular toxicity. Proc Natl Acad Sci USA
with myopathy, Paget disease of bone and frontotemporal dementia: 2009; 106: 7607–12.
review of a unique disorder. Biochim Biophys Acta 2008; 140 Li Y, Ray P, Rao EJ, et al. A Drosophila model for TDP-43
1782: 744–48. proteinopathy. Proc Natl Acad Sci USA 2010; 107: 3169–74.
119 Custer SK, Neumann M, Lu H, Wright AC, Taylor JP. Transgenic 141 Wils H, Kleinberger G, Janssens J, et al. TDP-43 transgenic mice
mice expressing mutant forms VCP/p97 recapitulate the full develop spastic paralysis and neuronal inclusions characteristic of
spectrum of IBMPFD including degeneration in muscle, brain and ALS and frontotemporal lobar degeneration. Proc Natl Acad Sci USA
bone. Hum Mol Genet 2010; 19: 1741–55. 2010; 107: 3858–63.
120 Vance C, Al-Chalabi A, Ruddy D, et al. Familial amyotrophic lateral 142 Tatom JB, Wang DB, Dayton RD, et al. Mimicking aspects of
sclerosis with frontotemporal dementia is linked to a locus on frontotemporal lobar degeneration and Lou Gehrig’s disease in rats
chromosome 9p13.2-21.3. Brain 2006; 129: 868–76. via TDP-43 overexpression. Mol Ther 2009; 17: 607–13.
121 Valdmanis PN, Dupre N, Bouchard JP, et al. Three families with 143 Zhou H, Huang C, Chen H, et al. transgenic rat model of
amyotrophic lateral sclerosis and frontotemporal dementia with neurodegeneration caused by mutation in the TDP gene.
evidence of linkage to chromosome 9p. Arch Neurol 2007; PLoS Genet 2010; 6: e1000887.
64: 240–45. 144 D’Ambrogio A, Buratti E, Stuani C, et al. Functional mapping of the
122 Morita M, Al-Chalabi A, Andersen PM, et al. A locus on interaction between TDP-43 and hnRNP A2 in vivo.
chromosome 9p confers susceptibility to ALS and frontotemporal Nucleic Acids Res 2009; 37: 4116–26.
dementia. Neurology 2006; 66: 839–44. 145 Johnson BS, Snead D, Lee JJ, McCaffery JM, Shorter J, Gitler AD.
123 Luty AA, Kwok JB, Thompson EM, et al. Pedigree with TDP-43 is intrinsically aggregation-prone, and amyotrophic lateral
frontotemporal lobar degeneration – motor neuron disease and sclerosis-linked mutations accelerate aggregation and increase
Tar DNA binding protein-43 positive neuropathology: genetic toxicity. J Biol Chem 2009; 284: 20329–39.
linkage to chromosome 9. BMC Neurol 2008; 8: 32. 146 Barmada SJ, Skibinski G, Korb E, Rao EJ, Wu JY, Finkbeiner S.
124 Le Ber I, Camuzat A, Berger E, et al. Chromosome 9p-linked Cytoplasmic mislocalization of TDP-43 is toxic to neurons and
families with frontotemporal dementia associated with motor enhanced by a mutation associated with familial amyotrophic
neuron disease. Neurology 2009; 72: 1669–76. lateral sclerosis. J Neurosci 2010; 30: 639–49.
125 Gijselinck I, Engelborghs S, Maes G, et al. Identification of 2 loci at 147 Wegorzewska I, Bell S, Cairns NJ, Miller TM, Baloh RH. TDP-43
chromosomes 9 and 14 in a multiplex family with frontotemporal mutant transgenic mice develop features of ALS and
lobar degeneration and amyotrophic lateral sclerosis. Arch Neurol frontotemporal lobar degeneration. Proc Natl Acad Sci USA 2009;
2010; 67: 606–16. 106: 18809–14.
126 Momeni P, Schymick J, Jain S, et al. Analysis of IFT74 as a candidate 148 Bertolotti A, Lutz Y, Heard DJ, Chambon P, Tora L. hTAF(II)68,
gene for chromosome 9p-linked ALS-FTD. BMC Neurol 2006; 6: 44. a novel RNA/ssDNA-binding protein with homology to the
127 van Es MA, Veldink JH, Saris CG, et al. Genome-wide association pro-oncoproteins TLS/FUS and EWS is associated with both
study identifies 19p13.3 (UNC13A) and 9p21.2 as susceptibility loci for TFIID and RNA polymerase II. EMBO J 1996; 15: 5022–31.
sporadic amyotrophic lateral sclerosis. Nat Genet 2009; 41: 1083–87. 149 Law WJ, Cann KL, Hicks GG. TLS, EWS and TAF15: a model for
128 Laaksovirta H, Peuralinna T, Schymick JC, et al. Chromosome 9p21 transcriptional integration of gene expression.
in amyotrophic lateral sclerosis in Finland: a genome-wide Brief Funct Genomic Proteomic 2006; 5: 8–14.
association study. Lancet Neurol 2010; published online Aug 31. 150 Lee BJ, Cansizoglu AE, Suel KE, Louis TH, Zhang Z, Chook YM.
DOI:10.1016/S1474-4422(10)70184-8. Rules for nuclear localization sequence recognition by karyopherin
129 Shatunov A, Mok K, Newhouse S, et al. Chromosome 9p21 in beta 2. Cell 2006; 126: 54–58.
sporadic amyotrophic lateral sclerosis in the UK and seven other 151 Zakaryan RP, Gehring H. Identification and characterization of the
countries: a genome-wide association study. Lancet Neurol 2010; nuclear localization/retention signal in the EWS proto-oncoprotein.
published online Aug 31. DOI:10.1016/S1474-4422(10)70197-6. J Mol Biol 2006; 363: 27–38.
130 Van Deerlin VM, Sleiman PM, Martinez-Lage M, et al. Common 152 Dormann D, Rodde R, Edbauer D, et al. ALS-associated fused in
variants at 7p21 are associated with frontotemporal lobar sarcoma (FUS) mutations disrupt transportin-mediated nuclear
degeneration with TDP-43 inclusions. Nat Genet 2010; 42: 234–39. import. EMBO J 2010; 29: 2841–57.
131 Ayala YM, Misteli T, Baralle FE. TDP-43 regulates retinoblastoma 153 Andersson MK, Stahlberg A, Arvidsson Y, et al. The multifunctional
protein phosphorylation through the repression of cyclin-dependent FUS, EWS and TAF15 proto-oncoproteins show cell type-specific
kinase 6 expression. Proc Natl Acad Sci USA 2008; 105: 3785–89. expression patterns and involvement in cell spreading and stress
132 Iguchi Y, Katsuno M, Niwa J, et al. TDP-43 depletion induces response. BMC Cell Biol 2008; 9: 37.
neuronal cell damage through dysregulation of Rho family 154 Zinszner H, Sok J, Immanuel D, Yin Y, Ron D. TLS (FUS) binds
GTPases. J Biol Chem 2009; 284: 22059–66. RNA in vivo and engages in nucleo-cytoplasmic shuttling.
133 Kabashi E, Lin L, Tradewell ML, et al. Gain and loss of function of J Cell Sci 1997; 110: 1741–50.
ALS-related mutations of TARDBP (TDP-43) cause motor deficits in 155 Janknecht R. EWS-ETS oncoproteins: the linchpins of Ewing
vivo. Hum Mol Genet 2010; 19: 671–83. tumors. Gene 2005; 363: 1–14.

1006 www.thelancet.com/neurology Vol 9 October 2010


Review

156 Fujii R, Okabe S, Urushido T, et al. The RNA binding protein TLS is 180 Cairns NJ, Perry RH, Jaros E, et al. Patients with a novel
translocated to dendritic spines by mGluR5 activation and regulates neurofilamentopathy: dementia with neurofilament inclusions.
spine morphology. Curr Biol 2005; 15: 587–93. Neurosci Lett 2003; 341: 177–80.
157 Fujii R, Takumi T. TLS facilitates transport of mRNA encoding an 181 Neumann M, Roeber S, Kretzschmar HA, Rademakers R, Baker M,
actin-stabilizing protein to dendritic spines. J Cell Sci 2005; Mackenzie IR. Abundant FUS-immunoreactive pathology in
118: 5755–65. neuronal intermediate filament inclusion disease. Acta Neuropathol
158 Hicks GG, Singh N, Nashabi A, et al. Fus deficiency in mice results 2009; 118: 605–16.
in defective B-lymphocyte development and activation, high levels 182 Doi H, Koyano S, Suzuki Y, Nukina N, Kuroiwa Y. The RNA-binding
of chromosomal instability and perinatal death. Nat Genet 2000; protein FUS/TLS is a common aggregate-interacting protein in
24: 175–79. polyglutamine diseases. Neurosci Res 2010; 66: 131–33.
159 Kuroda M, Sok J, Webb L, et al. Male sterility and enhanced 183 Woulfe J, Gray DA, Mackenzie IR. FUS-immunoreactive
radiation sensitivity in TLS(-/-) mice. EMBO J 2000; 19: 453–62. intranuclear inclusions in neurodegenerative disease.
160 Neumann M, Rademakers R, Roeber S, Baker M, Kretzschmar HA, Brain Pathol 2010; 20: 589–97.
Mackenzie IR. A new subtype of frontotemporal lobar degeneration 184 Lai SL, Abramzon Y, Schymick JC, et al. FUS mutations in sporadic
with FUS pathology. Brain 2009; 132: 2922–31. amyotrophic lateral sclerosis. Neurobiol Aging 2010; published
161 Suzuki N, Aoki M, Warita H, et al. FALS with FUS mutation in online Feb 6. DOI:10.1016/j.neurobiolaging.2009.12.020.
Japan, with early onset, rapid progress and basophilic inclusion. 185 Groen EJ, van Es MA, van Vught PW, et al. FUS mutations in
J Hum Genet 2010; 55: 252–54. familial amyotrophic lateral sclerosis in the Netherlands.
162 Roeber S, Mackenzie IR, Kretzschmar HA, Neumann M. TDP-43- Arch Neurol 2010; 67: 224–30.
negative FTLD-U is a significant new clinico-pathological subtype of 186 Drepper C, Herrmann T, Wessig C, Beck M, Sendtner M.
FTLD. Acta Neuropathol 2008; 116: 147–57. C-terminal FUS/TLS mutations in familial and sporadic ALS in
163 Mackenzie IR, Foti D, Woulfe J, Hurwitz TA. Atypical Germany. Neurobiol Aging 2009; published online Dec 9.
frontotemporal lobar degeneration with ubiquitin-positive, TDP-43- DOI:10.1016/j.neurobiolaging.2009.11.017.
negative neuronal inclusions. Brain 2008; 131: 1282–93. 187 DeJesus-Hernandez M, Kocerha J, Finch N, et al. De novo
164 Seelaar H, Klijnsma KY, de Koning I, et al. Frequency of ubiquitin truncating FUS gene mutation as a cause of sporadic amyotrophic
and FUS-positive, TDP-43-negative frontotemporal lobar lateral sclerosis. Hum Mutat 2010; 31: E1377–89.
degeneration. J Neurol 2010; 257: 747–53. 188 Damme PV, Goris A, Race V, et al. The occurrence of mutations in
165 Urwin H, Josephs KA, Rohrer JD, et al. FUS pathology defines the FUS in a Belgian cohort of patients with familial ALS. Eur J Neurol
majority of tau- and TDP-43-negative frontotemporal lobar 2010; 17: 754–56.
degeneration. Acta Neuropathol 2010 120: 33–41. 189 Corrado L, Del Bo R, Castellotti B, et al. Mutations of FUS gene in
166 Josephs KA, Whitwell JL, Parisi JE, et al. Caudate atrophy on MRI is sporadic amyotrophic lateral sclerosis. J Med Genet 2010; 47: 190–94.
a characteristic feature of FTLD-FUS. Eur J Neurol 2010 17: 969–75. 190 Chio A, Restagno G, Brunetti M, et al. Two Italian kindreds with
167 Yokota O, Tsuchiya K, Terada S, et al. Basophilic inclusion body familial amyotrophic lateral sclerosis due to FUS mutation.
disease and neuronal intermediate filament inclusion disease: a Neurobiol Aging 2009; 30: 1272–75.
comparative clinicopathological study. Acta Neuropathol 2008; 191 Belzil VV, Valdmanis PN, Dion PA, et al. Mutations in FUS cause
115: 561–75. FALS and SALS in French and French Canadian populations.
168 Munoz-Garcia D, Ludwin SK. Classic and generalized variants of Neurology 2009; 73: 1176–79.
Pick’s disease: a clinicopathological, ultrastructural, and 192 Waibel S, Neumann M, Rabe M, Meyer T, Ludolph AC. Novel
immunocytochemical comparative study. Ann Neurol 1984; 16: 467–80. missense and truncating mutations in FUS/TLS in familial ALS.
169 Tsuchiya K, Matsunaga T, Aoki M, et al. Familial amyotrophic lateral Neurology 2010; 75: 815–17.
sclerosis with posterior column degeneration and basophilic 193 Kobayashi Z, Tsuchiya K, Arai T, et al. Occurrence of basophilic
inclusion bodies: a clinical, genetic and pathological study. inclusions and FUS-immunoreactive neuronal and glial inclusions
Clin Neuropathol 2001; 20: 53–59. in a case of familial amyotrophic lateral sclerosis. J Neurol Sci 2010;
170 Aizawa H, Kimura T, Hashimoto K, Yahara O, Okamoto K, 293: 6–11.
Kikuchi K. Basophilic cytoplasmic inclusions in a case of sporadic 194 Gardiner M, Toth R, Vandermoere F, Morrice NA, Rouse J.
juvenile amyotrophic lateral sclerosis. J Neurol Sci 2000; 176: 109–13. Identification and characterization of FUS/TLS as a new target of
171 Matsumoto S, Kusaka H, Murakami N, Hashizume Y, Okazaki H, ATM. Biochem J 2008; 415: 297–307.
Hirano A. Basophilic inclusions in sporadic juvenile amyotrophic 195 Mayya V, Lundgren DH, Hwang SI, et al. Quantitative
lateral sclerosis: an immunocytochemical and ultrastructural study. phosphoproteomic analysis of T cell receptor signaling reveals
Acta Neuropathol 1992; 83: 579–83. system-wide modulation of protein-protein interactions. Sci Signal
172 Nelson JS, Prensky AL. Sporadic juvenile amyotrophic lateral 2009; 2: ra46.
sclerosis. A clinicopathological study of a case with neuronal 196 Ong SE, Mittler G, Mann M. Identifying and quantifying in vivo
cytoplasmic inclusions containing RNA. Arch Neurol 1972; 27: 300–06. methylation sites by heavy methyl SILAC. Nat Methods 2004; 1: 119–26.
173 Fujita K, Ito H, Nakano S, Kinoshita Y, Wate R, Kusaka H. 197 Rappsilber J, Friesen WJ, Paushkin S, Dreyfuss G, Mann M. Detection
Immunohistochemical identification of messenger RNA-related of arginine dimethylated peptides by parallel precursor ion scanning
proteins in basophilic inclusions of adult-onset atypical motor mass spectrometry in positive ion mode. Anal Chem 2003; 75: 3107–14.
neuron disease. Acta Neuropathol 2008; 116: 439–45. 198 Bedford MT, Clarke SG. Protein arginine methylation in mammals:
174 Kusaka H, Matsumoto S, Imai T. Adult-onset motor neuron disease who, what, and why. Mol Cell 2009; 33: 1–13.
with basophilic intraneuronal inclusion bodies. Clin Neuropathol 199 Allemand E, Guil S, Myers M, Moscat J, Caceres JF, Krainer AR.
1993; 12: 215–18. Regulation of heterogenous nuclear ribonucleoprotein A1 transport
175 Hamada K, Fukazawa T, Yanagihara T, et al. Dementia with ALS by phosphorylation in cells stressed by osmotic shock.
features and diffuse Pick body-like inclusions (atypical Pick’s Proc Natl Acad Sci USA 2005; 102: 3605–10.
disease?). Clin Neuropathol 1995; 14: 1–6. 200 Deng HX, Zhai H, Bigio EH, et al. FUS-immunoreactive inclusions
176 Ishihara K, Araki S, Ihori N, Shiota J, Kawamura M, Nakano I. An are a common feature in sporadic and non-SOD1 familial
autopsy case of frontotemporal dementia with severe dysarthria and amyotrophic lateral sclerosis. Ann Neurol 2010; 67: 739–48.
motor neuron disease showing numerous basophilic inclusions. 201 Maruyama H, Morino H, Ito H, et al. Mutations of optineurin in
Neuropathology 2006; 26: 447–54. amyotrophic lateral sclerosis. Nature 2010; 465: 223–26.
177 Munoz DG, Neumann M, Kusaka H, et al. FUS pathology in 202 Foulds P, McAuley E, Gibbons L, et al. TDP-43 protein in plasma may
basophilic inclusion body disease. Acta Neuropathol 2009; 118: 617–27. index TDP-43 brain pathology in Alzheimer’s disease and
178 Cairns NJ, Grossman M, Arnold SE, et al. Clinical and frontotemporal lobar degeneration. Acta Neuropathol 2008; 116: 141–46.
neuropathologic variation in neuronal intermediate filament 203 Steinacker P, Hendrich C, Sperfeld AD, et al. TDP-43 in cerebrospinal
inclusion disease. Neurology 2004; 63: 1376–84. fluid of patients with frontotemporal lobar degeneration and
179 Josephs KA, Holton JL, Rossor MN, et al. Neurofilament inclusion amyotrophic lateral sclerosis. Arch Neurol 2008; 65: 1481–87.
body disease: a new proteinopathy? Brain 2003; 126: 2291–303.

www.thelancet.com/neurology Vol 9 October 2010 1007

Anda mungkin juga menyukai