DataPDF Available

Liscic2017 ALSandFTDInsigIntoDiseaseMech

Authors:
  • Sachsenklinik Gmbh
Contents lists available at ScienceDirect
European Journal of Pharmacology
journal homepage: www.elsevier.com/locate/ejphar
Full length article
Als and Ftd: Insights into the disease mechanisms and therapeutic targets
Rajka M. Liscic
MD, PhD, Ass. Professor, EUREGIO KLINIK Albert-Schweitzer Straße GmbH, Nordhorn, Germany and Department of Anatomy and Neuroscience, School of Medicine,
University of Osijek, Croatia
ARTICLE INFO
Keywords:
ALS
Dementia
FTD
Genetics
Motor neuron disease
TDP-43
Riluzole
Stem cells
ABSTRACT
Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) are neurodegenerative dis-
orders, related by signs of deteriorating motor and cognitive functions, and short survival. The causes are still
largely unknown and no eective treatment currently exists. It has been shown that FTLD may coexist with ALS.
The overlap between ALS and frontotemporal dementia (FTD), the clinical syndrome associated with FTLD,
occurs at clinical, genetic, and pathological levels. The hallmark proteins of the pathognomonic inclusions are
SOD-1, TDP-43 or FUS, rarely the disease is caused by mutations in the respective genes. Frontotemporal lobar
degenerations (FTLD) is genetically, neuropathologically and clinically heterogeneous and may present with
behavioural, language and occasionally motor disorder, respectively. Almost all cases of ALS, as well as tau-
negative FTLD share a common neuropathology, neuronal and glial inclusion bodies containing abnormal TDP-
43 protein, collectively called TDP-43 proteinopathy. Recent discoveries in genetics (e.g. C9orf72 hexanucleo-
tide expansion) and the subsequent neuropathological characterization have revealed remarkable overlap be-
tween ALS and FTLD-TDP indicating common pathways in pathogenesis. For ALS, an anti-glutamate agent ri-
luzole may be oered to slow disease progression (Level A), and a promising molecule, arimoclomol, is currently
in clinical trials. Other compounds, however, are being trailed and some have shown encouraging results. As new
therapeutic approaches continue to emerge by targeting SOD1, TDP-43, or GRN, we present some advances that
are being made in our understanding of the molecular mechanisms of these diseases, which together with gene
and stem cell therapies may translate into new treatment options.
1. Introduction
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia
(FTD) are related clinical phenotypes, which are characterized by de-
cline in motor and cognitive functions, and short survival. ALS is the
most common adult-onset motor neuron disease, characterized by the
progressive, irreversible motor neuron loss leading to denervation
atrophy of muscles and death by respiratory failure (Brookset al.,
2000). Both diseases are invariably fatal after only a few years (Brooks
et al., 2000). The causes of the more frequent sporadic forms remain
unknown and no eective treatment currently exists. However, an anti-
glutamate agent Riluzole has shown modest benet and is the only drug,
so far, to receive US Food and Drug Administration (FDA) approval for
treatment of ALS (Miller et al., 1999). Also a Phase II/III randomised,
placebo-controlled trial of arimoclomol (BRX-345) in familial ALS with
SOD-1 mutation is underway (NCT00706147; www.ClinicalTrials.gov).
Arimoclomol, is a hydroxylamine derivative, a group of compounds
which have unique properties as co-inducers of heat shock protein ex-
pression, but only under conditions of cellular stress which is involved
in the ethiopathogenesis of multiple neurodegenerative diseases
including ALS (Kalmar et al., 2014). Treatment with arimoclomol has
been reported to improve survival and muscle function in dierent
mouse models of motor neuron disease including ALS and in mutant
superoxide dismutase 1 (SOD1) mice.
The small molecule arimoclomol may reduce the levels of protein
aggregates in motor nerves, a possible cause of ALS, by boosting ex-
pression of the heat shock proteins Hsp 70 and Hsp 90 which help
newly synthesised proteins to properly fold (www.alstdi.org;(Scotter
et al., 2015). Other compounds, including ceftriaxone and dexprami-
pexole, have been promoted and show encouraging results at phase II
clinical trials.
Ceftriaxone, belongs to a third generation of cafalosporine anti-
biotics, which can penetrate the blood brain barrier (BBB) and enter the
CNS. In addition to bactericidal action, ceftriaxone has been found in
various animal models to exert neuroprotective action caused by in-
creased expression of the astrocytic glutamate transporter, EAAT2 in
humans and GLT1 glutamate transporter in rodents. Glutamate trans-
porter EAAT2 increases the clearance of synaptic glutamate and pro-
tects neurons from glutamate-mediated excitotoxicity which is thought
to be a factor in their pathogenesis of ALS (Jagadapillai et al., 2014). In
http://dx.doi.org/10.1016/j.ejphar.2017.10.012
Received 27 October 2016; Received in revised form 6 October 2017; Accepted 9 October 2017
E-mail address: rajka.liscic@gmail.com.
European Journal of Pharmacology xxx (xxxx) xxx–xxx
0014-2999/ © 2017 Published by Elsevier B.V.
Please cite this article as: Liscic, R.M., European Journal of Pharmacology (2017), http://dx.doi.org/10.1016/j.ejphar.2017.10.012
ALS animal models, as well as in human post-mortem tissue, decreased
expression of glutamate transporter EAAT2 has been demonstrated.
Dexpramipexole is the enantiomer of pramipexole that is sig-
nicantly less eective dopamine receptor agonist but shows better
neuroprotective action. Dexpramipexole has been shown to inhibit the
opening of the mitochondrial permeability transition pore (PTP), im-
proving mitochondrial function and conferring signicant protection to
neurons exposed to free radicals under cellular stress (Vieira et al.,
2014).
Unfortunately, neither of these compounds showed ecacy at stage
III clinical trials (Cudkowicz et al., 2014, 2013), most likely due to
already well-established disease. Stem cell therapies are emerging as
potential disease modulating therapies in autoimmune diseases e.g.
rheumatoid arthritis, multiple sclerosis, and lupus (Mazzini et al.,
2010). Preclinical studies have shown that neurotrophic growth factors
(NTFs) extend the survival of motor neurons in ALS. Recently, an Israeli
research group developed a culture-based method for inducing me-
senchymal stem cells (MSCs) to secrete neurotrophic growth factors
(NTFs). These MSC-NTF cells have been shown to be protective in an-
imal models of neurodegenerative diseases. They successfully ad-
ministered autologous MSC-NTF cells, derived from bone marrow
samples given by participants (26 ALS patients) in the trial, via in-
trathecal (i.t) or intramuscular (i.m.) administration which proved to be
safe and provided indications of possible clinical benets. These ex-
periments are to be tested in upcoming clinical trials (Petrou et al.,
2016). This study showed that implanting stem cells that produce
neurotrophic factors (MSC-NTF) into either muscle or the intrathecal
space, or both, were shown to be safe and associated with only mild
adverse eects, principally headache and fever. This is the rst study in
which stem cells that have been modied to secrete neurotrophic fac-
tors have been implemented in patients with ALS. These results, how-
ever, should be treated with caution, since the study included relatively
small group of patients and the trial did not include a placebo com-
parator.
Immunological therapies originally proved more promising, with a
Phase III antibody-Nogo study. The Nogo A is one of the three isoforms
of the Nogo protein that acts as a neurite outgrowth inhibitor of the
Nogo protein, encoded by the RTN4 gene (Pernet and Schwab, 2012).
However, although the study has been completed (NCT00406016,
www.ClinicalTrials.gov), no results have been published. For more on
clinical trials, please refer to www.ClinicalTrials.gov.
In FTD, which typically presents with either behavioural abnorm-
alities or language dysfunction, a considerable number of patients also
develop muscle weakness and wasting that is typical for ALS (Strong,
2008; Strong et al., 2009) or, vice versa, ALS patients may develop
impaired executive dysfunction and memory decline (Lomen-Hoerth
et al., 2002; Lomen-Hoerth et al., 2003). The presence of fronto-
temporal impairment in ALS predicts a shorter survival time (Hodges
et al., 2003) and behavioural and functional impairment may decline
independently of motor function (De Silva et al., 2015; Rohrer et al.,
2015).
With regard to treatments for FTD, a lot of symptomatic treatments
have been reported so far. Open-label studies of anticholinesterase
medicines have been negative and, in some cases they may even ex-
acerbate behavioural symptoms (Devenney, Vucic, Hodges, and
Kiernan, 2015). Similarly, a memantine study showed no eective
treatment in a recent randomized placebo controlled trial (Boxer et al.,
2012). The selective serotonin reuptake inhibitors and antipsychotic
therapies are generally considered helpful in the management of mood
and behavioural features in individual patients despite lack of evidence
(Devenney et al., 2015). Tauopathy, however, has been a focus for re-
search and the development of potential disease-modifying treatments
(Devenney et al., 2015). According to the latest study on tau-transgenic
mice, methylene blue substance, a drug in Phase III clinical trials stops
decline in its tracks- but only when given at the earliest stages
(Hochgrafe et al., 2015). This TRx-237-007 Phase 3 clinical trial
conducted in 220 subjects with bvFTD was stopped, as it failed to reach
its co-primary endpoints (TauRx Therapeutics Ltd. Sep 2016 conference
news; company press release).
The incidence of ALS in Europe is 2.16 per 100,000 (Logroscino
et al., 2010), while for FTD the incidence is 3.5 per 100,000 (Mercy
et al., 2008). Here we present a current understanding of molecular
causes of ALS and FTLD, in order to better understand pathogenetic
mechanisms of both diseases as new design for clinical trials emerges in
patients with ALS and FTD.
2. Genetics
A growing number of ALS-causing genes have been identied re-
cently. These are now under investigation as these may shed some light
on pathogenesis. Four major ALS-associated genes are: superoxide dis-
mutase 1 (SOD1), the rst genetic cause to be identied, TAR DNA-
binding protein (TARDBP), fusion in malignant liposarcoma/
translocation in liposarcoma (FUS/TLS), and the most common
chromosome 9 open reading frame 72 (C9orf72).
Identication of disease linked mutations in TDP-43 and FUS/TLS
highlight dysfunctions in RNA metabolism as a common pathogenic
pathway in both ALS and FTD. TDP-43 and FUS/TLS have similar
structures and are involved in several RNA processing steps. TDP-43
binds to more than 6000 RNA targets and acts as a transcriptional re-
pressor through its direct binding to DNA. It is involved in splicing,
microRNA biogenesis, RNA transport and translation, and stress granule
formation. However, FUS/TLS can bind to a single or double-stranded
DNA and RNA. FUS/TLS interacts with RNA polymerase II and pro-
motes or represses the transcription of distinct genes and aects
Fig. 1. Pathology in cases of frontotemporal lobar degeneration-motor neuron disease
(FTLD-ALS) with C9orf72 expansion. The gure shows ubiquitin-positive, TDP-43-nega-
tive neuronal cytoplasmic inclusions (arrows) in the granule cells of the cerebellum (p62
immunohistochemistry). Scale bars: 20 micrometres. Courtesy of N.J. Cairns, Washington
University in St. Louis, USA.
Fig. 2. Pathology in cases of frontotemporal lobar degeneration-motor neuron disease
(FTLD-ALS) with C9orf72 expansion. The gure shows the cerebellar inclusions are la-
belled with anti-dipeptide repeat antibodies (poly-GA immunohistochemistry). Scale bars:
20 micrometres. Courtesy of N.J. Cairns, Washington University in St. Louis, USA.
R.M. Liscic European Journal of Pharmacology xxx (xxxx) xxx–xxx
2
splicing. Both, TDP-43 and FUS/TLS can modify the function of stress
granules and regulate synaptic function. When nuclear TDP is depleted,
it aects not only the expression of TDP-43, but also FUS/TLS, glial
transporter EAAT2, amyloid-beta precursor protein, presenilin, hun-
tingtin, multiple ataxins, alpha-synuclein, and progranulin. The most
aected molecular species is pre-mRNA (Ling et al., 2013).
In addition to neuronal inclusion bodies, both TDP-43 and FUS/TLS
aggregates may be found in glial cells. Another mutual characteristics of
ALS/FTP is a reactive gliosis (Radford et al., 2015). Reactive gliosis is
an unspecic response to noxious stimuli within brain and is char-
acterized by microglial proliferation and astrocytic hypertrophy. ALS-
FTP-induced reactive gliosis is the most prominent in the brain areas
showing neuronal loss and inclusion pathology. The pathologic function
of glia can further contribute to neurodegeneration in ALS/FTP by re-
leasing glia-derived neurotoxic compounds and by reducing the clear-
ance potential of glia. In addition to decreased phagocytic activity by
reactive glial cells, the reduced vascular and glymphatic ow which is
controlled by glia participates to disease spreading and progression (Ng
et al., 2015).
In 2011, the discovery of the hexanucleotide repeat expansion in the
C9orf72 gene was revealed as the most frequent genetic cause of both
ALS and FTD (DeJesus-Hernandez et al., 2011)(Renton et al., 2011). An
expanded hexanucleotide, G
4
C
2,
repeat in C9orf72, is pathogenic at
greater than 30 repeats (Fratta et al., 2015), with most patients having
expansions > 500 repeats. The characteristic pathological ndings in
both ALS and FTLD are focal neuronal loss and gliosis and neuronal
cytoplasmic inclusion bodies containing TDP-43. Of the TDP-43 pro-
teinopathies, C9orf72 mutations are unique in that many inclusion
bodies also contain dipeptide repeat (DPR) proteins. Possible molecular
mechanisms of neurodegeneration include loss or gain in function of
the C9orf72 protein (Mizielinska et al., 2014; Rohrer et al., 2015). Loss
of protein function reects reduced levels of C9orf72 in brain and in
vitro studies reveal a role for C9orf72 protein in endosomal and au-
tophagic pathways (Mizielinska et al., 2014). A toxic gain of function is
supported by the presence of both repeat RNA and protein aggregates in
post-mortem brain. Repeat RNA aggregates, or RNA foci, have been
shown to sequester proteins involved in RNA splicing, editing, nuclear
export, and nuclear function. An alternative-RNA-mediated mechanism
to RNA binding protein is associated with non-TGA (RAN) translation
which produces aberrant peptides or dipeptide polymers that form a
proportion of inclusion bodies in brain tissue (Zu et al., 2011). In
combating gain-of-function toxicity, oligonucleotides targeting C9orf72
show some success (Mizielinska and Isaacs, 2014).
Oligonucleotide-based antisense techniques represent to date one of
the more successful approaches to achieving suppression or elimination
of a genetic message. Classes of antisense oligonucleotide therapeutics
can be divided into: RNase-H-dependent, exon skipping antisense oli-
gonucleotides, siRNAs, anti-miRNA and miRNA mimics. RNase H-de-
pendent and exon skipping antisense nucleotides are single stranded,
chemically modied oligonucleotides. RNase-H-dependent oligonu-
cleotides bind to mRNA and reduce the gene expression by RNase-H
mediated cleavage of the target RNA and by inhibition of translation by
steric blockade of ribosomes, whereas exon skipping target intro-exon
junctions or splicing-regulatory elements. siRNA are small 2123 nu-
cleotide long chemically modied with an antisense strand that is
complementary to a sequence anywhere in the target RNA. Anti-mi-Rs
and miRNA mimics either antagonise or mimics the endogenous
miRNA, but less eciently than si RNAs. Oligonucleotides targeting
C9orf72 belong to RNase-H-dependent antisense nucleotides (Rosen
et al., 1993).
Three major ALS-associated genes, SOD1, which encodes Cu/Zn
superoxide dismutase-1 (Rosen et al., 1993), TARDBP, and FUS/TLS
have been identied with a growing number of ALS-causing genes.
These genes are now under investigation as providing promise for in-
creased understanding of pathogenesis of the disease (Liscic and
Breljak, 2011). Other genes, including angiogenin (ANG), the vesicle-
associated membrane protein-associated protein B (VAPB), senataxin
(SETX), and dynactin gene have been also identied in ALS patients.
Mutations in the FUS/TLS gene, located on chromosome 16, have been
identied as a causal gene for ~4% of FALS (~0.4% of all ALS)
(Kwiatkowski et al., 2009; Vance et al., 2009) and FTLD (Van
Langenhove et al., 2010). As with TDP-43, FUS is a predominantly
nuclear protein that is expressed at low levels in the cytoplasm (Lagier-
Tourenne and Cleveland, 2009). Although the phenotype associated
with FUS mutation is variable, most patients predominantly demon-
strate loss of lower motor neurons and short disease survival. Mutations
in the SOD1 were rst described in 1993 (Rosen et al., 1993). Since then
more than 120 dierent SOD1 mutations have been identied and
claimed to be responsible for 20% of FALS cases (Liscic and Breljak,
2011). In 2008, Gitcho and colleagues (Gitcho et al., 2008) and
Sreedharan and colleagues (Sreedharan et al., 2008) independently
reported pathogenic mutations in the TARDBP gene located on chro-
mosome 1 encoding TAR DNA-binding protein 43 (TDP-43), which
cause several neurodegenerative diseases such as FALS, sporadic ALS,
and FTLD. Their ndings support a direct role of TARDBP mutations in
neurodegeneration. TDP-43 is a 414-amino acid ubiquitously expressed
nuclear protein, which contains two highly conserved RNA-recognition
motifs (RRM1 and RRM2), a nuclear localisation signal at the N-ter-
minus, and a glycine-rich region mediating protein-protein interactions
at the C-terminus (Lagier-Tourenne and Cleveland, 2009). Pathological
C-terminal TDP-43 fragments of 25 kDa are ubiquitinated, hyperpho-
sphorylated and accumulated as cellular inclusions in neurons and glial
cells (Neumann et al., 2006). To explore the link between mutations in
TARDBP and the spectrum of TDP-43 proteinopathies TDP-43 mutant
transgenic mice models with clinical features of ALS and FTD have been
developed, however, with varying results (Wegorzewska et al., 2009;
Wils et al., 2010). Although it may be impossible to model accurately
one clinical feature of a human neurodegenerative disease in mice,
mouse models are useful in that they shed light on pathogenic me-
chanisms causing cell death and he clinical phenotypes associated with
both ALS and FTD; however, mouse models have had only limited
success in recapitulating the neuropathology of TDP-43 proteinopathy
(Ittner et al., 2015). The identication of SOD-1 mutations in FALS and
MAPT mutations in FTDP-17 (Rosen et al., 1993) prompted the gen-
eration of several mouse TARDBP mutants (Igaz et al., 2011). However,
none of these models is perfect. Mutations in GRN also result in TDP-43
neuropathology in humans, but knockout mice show little pathologi-
cally phosphorylated TDP-43 (Wils et al., 2012), thus indicating that
the link between gene defect and pathology is complex. Recently, the
rst (Hukema et al., 2014) repeat-expansion mouse model and
knockout mice was reported (Panda et al., 2013), but without a human-
related pathologic phenotype. The high prevalence of C9orf72 muta-
tions in familial FTD-ALS suggests that accurate mouse models of this
mutation will be necessary to facilitate drug discovery and develop-
ment.
3. Neuropathology of ALS and FTLD (frontotemporal lobar
degeneration)
Clinically and neuropathologically, ALS and FTD or its pathological
substrate called frontotemporal lobar degeneration (FTLD) may be seen
as two ends of a spectrum (Cairns et al., 2007). For example, TDP-43
proteinopathy characterizes most sporadic cases of both ALS and FTLD.
However, the signicance of TDP-43-immunoreactive inclusion bodies
in the pathogenesis of ALS-FTLD remains enigmatic (Buratti, 2015).
The neuropathology of ALS is characterized mainly by the abnormal
accumulation of insoluble proteins as inclusion bodies in the cytoplasm
of degenerating motor neurons (Lowe, 1994; Neumann et al., 2006).
Until recently, the specic biochemical composition of these neuronal
cytoplasmic inclusions (NCIs) was unknown, except that the abnormal
protein was ubiquitinated. These ubiquitin-immunoreactive (ub-ir)
NCIs are most common in surviving lower motor neurons and most
R.M. Liscic European Journal of Pharmacology xxx (xxxx) xxx–xxx
3
often appear as either lamentous skeins or compact round bodies
(Lowe, 1994).
ALS is accompanied by a wide range of neuropathological features
in which both cortical (upper motor neuron) and either brainstem
motor neurons or anterior horn cells (lower motor neuron) are involved
with the signature lesion: abnormal accumulation of aggregated pro-
teins in cytoplasm of degenerating motor neurons (Wong et al., 1995).
Until recently, the pathological proteins in these inclusion bodies of ALS
and FTL with ubiquitin-immunoreactive inclusions were largely. TDP-
43 was identied as the most frequent protein in the inclusions of both
ALS an FTLD (Liscic, 2015; Liscic, Grinberg, Zidar, Gitcho, and Cairns,
2008). TDP-43 is normally found within the nucleus but under patho-
logical conditions, TDP-43 is translocated to the cytoplasm where ab-
normal inclusion bodies are formed. Although most inclusions are seen
within neurons they may also be found in glial cells. Biochemical
analysis of puried NCIs indicates that pathologic TDP-43 is ubiquiti-
nated, hyperphosphrylated, and accumulates as abnormal C-terminally
fragments of 25 kDa (Neumann et al., 2006). TFP-43 proteinopathy
now constitutes the most frequent molecular pathology of FTLD; about
half of cases contain abnormal tau, called the tauopathies, and a re-
sidual group of about 5 per cent of cases are characterized aby ab-
normal inclusion bodies containing FUS, collectively called the FUS
proteinopathies. But it is not clear weather aggregation of TDP-43 is a
primary event in ALS pathogenesis or whether it is a by-product of
another pathological process. Phosphorylated 43 kDa TAR DNA-
binding protein (pTDP-43) intraneuronal inclusions in ALS are found
within the motor cortex, brainstem motor nuclei, cranial nerve nuclei V,
VII, and X-XII, and in spinal cord motor neurons. In cases with a
C9orf72 hexanucleotide expansion there may be a more orid TDP-43
molecular pathology (Brettschneider et al., 2014). A unique feature of
C9orf72 cases is the presence of dipeptide repeat proteins in many in-
clusion bodies that are TDP-43 negative (Al-Sarraj et al., 2011), Figs. 1
and 2.
4. Innovative perspectives and conclusion
Most ALS and FTLD cases are progressive, relatively frequent,
neurodegenerative diseases without a known cause. In familial ALS
(FALS) many causative gene defects have been described (for an up-to-
date list see: http://alsod.iop.kcl.ac.uk/). The only drug currently li-
censed for the symptomatic ALS treatment today is an anti/glutamate
agent Riluzole. Also, immunological therapies proved promising, with a
Phase III Nogo Antibody study and an investigational agent arimoclomol
currently in clinical trials. However, the Nogo antibody study showed
no encouraging results.
Recently, new emerging results were presented with stem cell
therapies as potential disease modulating therapies. The bone marrow
cells were turned into stem cells that secrete neurotrophic factor (NTF).
By implanting mesenchymal stem cells (MSC) that produce neuro-
trophic factor (MSC-NTF) in patients with ALS into either muscle or the
intrathecal space, or both, the study showed to be safe and associated
with only mild adverse eects, principally headache and fever. This is
the rst study in which stem cells that have been modied to secrete
neurotrophic factors have been tested in patients with ALS. These re-
sults, however, should be treated with caution since the study included
relatively small numbers of patients and the trial did not include a
placebo comparator.
Major discoveries, however, have been made in the recent past in
the genetics, biochemistry, and neuropathology of ALS and FTLD. The
C9orf72 hexanucleotide repeat expansion is the most common genetic
cause of both ALS and FTD; all of these cases display TDP-43.
Interestingly, multiple gene defects in C9orf72,TARDBP, and VCP all
produce TDP-43 proteinopathy with C9orf72 cases having unique pa-
thological features. Conversely, mutations in a single gene, MAPT,
produce a heterogeneous pathology, tauopathy, and mutations in FUS/
TLS are linked to FALS with FUS proteinopathy. The revised
classication of ALS and FTLD according to genetics and molecular
pathology is providing new insights into pathogenesis and potential
new targets for therapeutic intervention. Accompanying these ap-
proaches, gene (and eventually stem cell) therapies may provide some
relief to these, at present, incurable neurodegenerative diseases.
Acknowledgement
Figures were kindly provided by NJ Cairns, PhD, Washington
University School of Medicine, St. Louis, Missouri, U.S.A.
Disclosure
The author declares no competing conict of interest.
References
Al-Sarraj, S., King, A., Troakes, C., Smith, B., Maekawa, S., Bodi, I., Shaw, C.E., 2011. P62
positive, TDP-43 negative, neuronal cytoplasmic and intranuclear inclusions in the
cerebellum and hippocampus dene the pathology of C9orf72-linked FTLD and
MND/ALS. Acta Neuropathol. 122 (6), 691702. http://dx.doi.org/10.1007/s00401-
011-0911-2.
Boxer, A.L., Gold, M., Huey, E., Gao, F.B., Burton, E.A., Chow, T., Cummings, J., 2012.
Frontotemporal degeneration, the next therapeutic frontier: molecules and animal
models for frontotemporal degeneration drug development. Alzheimers Dement.
http://dx.doi.org/10.1016/j.jalz.2012.03.002.
Brettschneider, J., Arai, K., Del Tredici, K., Toledo, J.B., Robinson, J.L., Lee, E.B.,
Trojanowski, J.Q., 2014. TDP-43 pathology and neuronal loss in amyotrophic lateral
sclerosis spinal cord. Acta Neuropathol. 128 (3), 423437. http://dx.doi.org/10.
1007/s00401-014-1299-6.
Brooks, B.R., Miller, R.G., Swash, M., Munsat, T.L., World Federation of Neurology
Research Group on Motor Neuron, D, 2000. El escorial revisited: revised criteria for
the diagnosis of amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. Other Mot.
Neuron Disord. 1 (5), 293299.
Buratti, E., 2015. Functional signicance of TDP-43 mutations in disease. Adv. Genet 91,
153. http://dx.doi.org/10.1016/bs.adgen.2015.07.001.
Cairns, N.J., Bigio, E.H., Mackenzie, I.R., Neumann, M., Lee, V.M., Hatanpaa, K.J.,
Consortium for Frontotemporal Lobar, D, 2007. Neuropathologic diagnostic and
nosologic criteria for frontotemporal lobar degeneration: consensus of the
Consortium for frontotemporal lobar degeneration. Acta Neuropathol. 114 (1), 522.
http://dx.doi.org/10.1007/s00401-007-0237-2.
Cudkowicz, M.E., Titus, S., Kearney, M., Yu, H., Sherman, A., Schoenfeld, D., Ceftriaxone
Study, I, 2014. Safety and ecacy of ceftriaxone for amyotrophic lateral sclerosis: a
multi-stage, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 13
(11), 10831091. http://dx.doi.org/10.1016/S1474-4422(14)70222-4.
Cudkowicz, M.E., van den Berg, L.H., Shefner, J.M., Mitsumoto, H., Mora, J.S., Ludolph,
A., investigators, E., 2013. Dexpramipexole versus placebo for patients with amyo-
trophic lateral sclerosis (EMPOWER): a randomised, double-blind, phase 3 trial.
Lancet Neurol. 12 (11), 10591067. http://dx.doi.org/10.1016/S1474-4422(13)
70221-7.
De Silva, D., Hsieh, S., Caga, J., Leslie, F.V., Kiernan, M.C., Hodges, J.R., Burrell, J.R.,
2015. Motor function and behaviour across the ALS-FTD spectrum. Acta Neurol.
Scand. http://dx.doi.org/10.1111/ane.12471.
DeJesus-Hernandez, M., Mackenzie, I.R., Boeve, B.F., Boxer, A.L., Baker, M., Rutherford,
N.J., Rademakers, R., 2011. Expanded GGGGCC hexanucleotide repeat in noncoding
region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 72 (2),
245256. http://dx.doi.org/10.1016/j.neuron.2011.09.011.
Devenney, E., Vucic, S., Hodges, J.R., Kiernan, M.C., 2015. Motor neuron disease-fron-
totemporal dementia: a clinical continuum. Expert Rev. Neurother. 15 (5), 509522.
http://dx.doi.org/10.1586/14737175.2015.1034108.
Fratta, P., Polke, J.M., Newcombe, J., Mizielinska, S., Lashley, T., Poulter, M., Fisher,
E.M., 2015. Screening a UK amyotrophic lateral sclerosis cohort provides evidence of
multiple origins of the C9orf72 expansion (546 e541-547). Neurobiol. Aging 36 (1).
http://dx.doi.org/10.1016/j.neurobiolaging.2014.07.037.
Gitcho, M.A., Baloh, R.H., Chakraverty, S., Mayo, K., Norton, J.B., Levitch, D., Cairns,
N.J., 2008. TDP-43 A315T mutation in familial motor neuron disease. Ann. Neurol.
63 (4), 535538. http://dx.doi.org/10.1002/ana.21344.
Hochgrafe, K., Sydow, A., Matenia, D., Cadinu, D., Konen, S., Petrova, O., Mandelkow,
E.M., 2015. Preventive methylene blue treatment preserves cognition in mice ex-
pressing full-length pro-aggregant human Tau. Acta Neuropathol. Commun. 3, 25.
http://dx.doi.org/10.1186/s40478-015-0204-4.
Hodges, J.R., Davies, R., Xuereb, J., Kril, J., Halliday, G., 2003. Survival in fronto-
temporal dementia. Neurology 61 (3), 349354.
Hukema, R.K., Riemslagh, F.W., Melhem, S., van der Linde, H.C., Severijnen, L.A.,
Edbauer, D., van Swieten, J.C., 2014. A new inducible transgenic mouse model for
C9orf72-associated GGGGCC repeat expansion supports a gain-of-function me-
chanism in C9orf72-associated ALS and FTD. Acta Neuropathol. Commun. 2, 166.
http://dx.doi.org/10.1186/s40478-014-0166-y.
Igaz, L.M., Kwong, L.K., Lee, E.B., Chen-Plotkin, A., Swanson, E., Unger, T., Lee, V.M.,
2011. Dysregulation of the ALS-associated gene TDP-43 leads to neuronal death and
R.M. Liscic European Journal of Pharmacology xxx (xxxx) xxx–xxx
4
degeneration in mice. J. Clin. Invest 121 (2), 726738. http://dx.doi.org/10.1172/
JCI44867.
Ittner, L.M., Halliday, G.M., Kril, J.J., Gotz, J., Hodges, J.R., Kiernan, M.C., 2015. FTD
and ALStranslating mouse studies into clinical trials. Nat. Rev. Neurol. 11 (6),
360366. http://dx.doi.org/10.1038/nrneurol.2015.65.
Jagadapillai, R., Mellen, N.M., Sachleben, L.R., Jr, Gozal, E., 2014. Ceftriaxone preserves
glutamate transporters and prevents intermittent hypoxia-induced vulnerability to
brain excitotoxic injury. Plos One 9 (7), e100230. http://dx.doi.org/10.1371/
journal.pone.0100230.
Kalmar, B., Lu, C.H., Greensmith, L., 2014. The role of heat shock proteins in amyotrophic
lateral sclerosis: the therapeutic potential of arimoclomol. Pharmacol. Ther. 141 (1),
4054. http://dx.doi.org/10.1016/j.pharmthera.2013.08.003.
Kwiatkowski, T.J., Jr, Bosco, D.A., Leclerc, A.L., Tamrazian, E., Vanderburg, C.R., Russ,
C., Brown, R.H., Jr, 2009. Mutations in the FUS/TLS gene on chromosome 16 cause
familial amyotrophic lateral sclerosis. Science 323 (5918), 12051208. http://dx.doi.
org/10.1126/science.1166066.
Lagier-Tourenne, C., Cleveland, D.W., 2009. Rethinking ALS: the FUS about TDP-43. Cell
136 (6), 10011004. http://dx.doi.org/10.1016/j.cell.2009.03.006.
Ling, S.C., Polymenidou, M., Cleveland, D.W., 2013. Converging mechanisms in ALS and
FTD: disrupted RNA and protein homeostasis. Neuron 79 (3), 416438. http://dx.doi.
org/10.1016/j.neuron.2013.07.033.
Liscic, R.M., 2015. Molecular basis of ALS and FTD: implications for translational studies.
Arh. Hig. Rada Toksikol. 66 (4), 285290. http://dx.doi.org/10.1515/aiht-2015-66-
2679.
Liscic, R.M., Breljak, D., 2011. Molecular basis of amyotrophic lateral sclerosis. Prog.
Neuropsychopharmacol. Biol. Psychiatry 35 (2), 370372. http://dx.doi.org/10.
1016/j.pnpbp.2010.07.017.
Liscic, R.M., Grinberg, L.T., Zidar, J., Gitcho, M.A., Cairns, N.J., 2008. ALS and FTLD: two
faces of TDP-43 proteinopathy. Eur. J. Neurol. 15 (8), 772780. http://dx.doi.org/10.
1111/j.1468-1331.2008.02195.x.
Logroscino, G., Traynor, B.J., Hardiman, O., Chio, A., Mitchell, D., Swingler, R.J., Eurals,
2010. Incidence of amyotrophic lateral sclerosis in Europe. J. Neurol. Neurosurg.
Psychiatry 81 (4), 385390. http://dx.doi.org/10.1136/jnnp.2009.183525.
Lomen-Hoerth, C., Anderson, T., Miller, B., 2002. The overlap of amyotrophic lateral
sclerosis and frontotemporal dementia. Neurology 59 (7), 10771079.
Lomen-Hoerth, C., Murphy, J., Langmore, S., Kramer, J.H., Olney, R.K., Miller, B., 2003.
Are amyotrophic lateral sclerosis patients cognitively normal? Neurology 60 (7),
10941097.
Lowe, J., 1994. New pathological ndings in amyotrophic lateral sclerosis. J. Neurol. Sci.
124, Suppl, 3851.
Mazzini, L., Ferrero, I., Luparello, V., Rustichelli, D., Gunetti, M., Mareschi, K., Fagioli, F.,
2010. Mesenchymal stem cell transplantation in amyotrophic lateral sclerosis: a
Phase I clinical trial. Exp. Neurol. 223 (1), 229237. http://dx.doi.org/10.1016/j.
expneurol.2009.08.007.
Mercy, L., Hodges, J.R., Dawson, K., Barker, R.A., Brayne, C., 2008. Incidence of early-
onset dementias in Cambridgeshire, United Kingdom. Neurology 71 (19), 14961499.
http://dx.doi.org/10.1212/01.wnl.0000334277.16896.fa.
Miller, R.G., Rosenberg, J.A., Gelinas, D.F., Mitsumoto, H., Newman, D., Sut, R.,
Oppenheimer, E.A., 1999. practice parameter: the care of the patient with amyo-
trophic lateral sclerosis (an evidence-based review): report of the quality standards
subcommittee of the American academy of neurology: ALS practice parameters task
force. Neurology 52 (7), 13111323.
Mizielinska, S., Gronke, S., Niccoli, T., Ridler, C.E., Clayton, E.L., Devoy, A., Isaacs, A.M.,
2014. C9orf72 repeat expansions cause neurodegeneration in Drosophila through
arginine-rich proteins. Science 345 (6201), 11921194. http://dx.doi.org/10.1126/
science.1256800.
Mizielinska, S., Isaacs, A.M., 2014. C9orf72 amyotrophic lateral sclerosis and fronto-
temporal dementia: gain or loss of function? Curr. Opin. Neurol. 27 (5), 515523.
http://dx.doi.org/10.1097/WCO.0000000000000130.
Neumann, M., Sampathu, D.M., Kwong, L.K., Truax, A.C., Micsenyi, M.C., Chou, T.T., Lee,
V.M., 2006. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyo-
trophic lateral sclerosis. Science 314 (5796), 130133. http://dx.doi.org/10.1126/
science.1134108.
Ng, A.S., Rademakers, R., Miller, B.L., 2015. Frontotemporal dementia: a bridge between
dementia and neuromuscular disease. Ann. N. Y Acad. Sci. 1338, 7193. http://dx.
doi.org/10.1111/nyas.12638.
Panda, S.K., Wefers, B., Ortiz, O., Floss, T., Schmid, B., Haass, C., Kuhn, R., 2013. Highly
ecient targeted mutagenesis in mice using TALENs. Genetics 195 (3), 703713.
http://dx.doi.org/10.1534/genetics.113.156570.
Pernet, V., Schwab, M.E., 2012. The role of Nogo-A in axonal plasticity, regrowth and
repair. Cell Tissue Res 349 (1), 97104. http://dx.doi.org/10.1007/s00441-012-
1432-6.
Petrou, P., Gothelf, Y., Argov, Z., Gotkine, M., Levy, Y.S., Kassis, I., Karussis, D., 2016.
Safety and clinical eects of mesenchymal stem cells secreting neurotrophic factor
transplantation in patients with amyotrophic lateral sclerosis: results of phase 1/2
and 2a clinical trials. JAMA Neurol. 73 (3), 337344. http://dx.doi.org/10.1001/
jamaneurol.2015.4321.
Radford, R.A., Morsch, M., Rayner, S.L., Cole, N.J., Pountney, D.L., Chung, R.S., 2015.
The established and emerging roles of astrocytes and microglia in amyotrophic lateral
sclerosis and frontotemporal dementia. Front Cell Neurosci. 9, 414. http://dx.doi.
org/10.3389/fncel.2015.00414.
Renton, A.E., Majounie, E., Waite, A., Simon-Sanchez, J., Rollinson, S., Gibbs, J.R.,
Traynor, B.J., 2011. A hexanucleotide repeat expansion in C9ORF72 is the cause of
chromosome 9p21-linked ALS-FTD. Neuron 72 (2), 257268. http://dx.doi.org/10.
1016/j.neuron.2011.09.010.
Rohrer, J.D., Isaacs, A.M., Mizielinska, S., Mead, S., Lashley, T., Wray, S., Warren, J.D.,
2015. C9orf72 expansions in frontotemporal dementia and amyotrophic lateral
sclerosis. Lancet Neurol. 14 (3), 291301. http://dx.doi.org/10.1016/S1474-
4422(14)70233-9.
Rosen, D.R., Siddique, T., Patterson, D., Figlewicz, D.A., Sapp, P., Hentati, A., et al., 1993.
Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyo-
trophic lateral sclerosis. Nature 362 (6415), 5962. http://dx.doi.org/10.1038/
362059a0.
Scotter, E.L., Chen, H.J., Shaw, C.E., 2015. TDP-43 Proteinopathy and ALS: Insights into
Disease Mechanisms and Therapeutic Targets. Neurotherapeutics 12 (2), 352363.
http://dx.doi.org/10.1007/s13311-015-0338-x.
Sreedharan, J., Blair, I.P., Tripathi, V.B., Hu, X., Vance, C., Rogelj, B., Shaw, C.E., 2008.
TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science 319
(5870), 16681672. http://dx.doi.org/10.1126/science.1154584.
Strong, M.J., 2008. The syndromes of frontotemporal dysfunction in amyotrophic lateral
sclerosis. Amyotroph. Lateral Scler. 9 (6), 323338. http://dx.doi.org/10.1080/
17482960802372371.
Strong, M.J., Grace, G.M., Freedman, M., Lomen-Hoerth, C., Woolley, S., Goldstein, L.H.,
Figlewicz, D., 2009. Consensus criteria for the diagnosis of frontotemporal cognitive
and behavioural syndromes in amyotrophic lateral sclerosis. Amyotroph. Lateral
Scler. 10 (3), 131146.
Van Langenhove, T., van der Zee, J., Sleegers, K., Engelborghs, S., Vandenberghe, R.,
Gijselinck, I., Van Broeckhoven, C., 2010. Genetic contribution of FUS to fronto-
temporal lobar degeneration. Neurology 74 (5), 366371. http://dx.doi.org/10.
1212/WNL.0b013e3181ccc732.
Vance, C., Rogelj, B., Hortobagyi, T., De Vos, K.J., Nishimura, A.L., Sreedharan, J., Shaw,
C.E., 2009. Mutations in FUS, an RNA processing protein, cause familial amyotrophic
lateral sclerosis type 6. Science 323 (5918), 12081211. http://dx.doi.org/10.1126/
science.1165942.
Vieira, F.G., LaDow, E., Moreno, A., Kidd, J.D., Levine, B., Thompson, K., Perrin, S., 2014.
Dexpramipexole is ineective in two models of ALS related neurodegeneration. Plos
One 9 (12), e91608. http://dx.doi.org/10.1371/journal.pone.0091608.
Wegorzewska, I., Bell, S., Cairns, N.J., Miller, T.M., Baloh, R.H., 2009. TDP-43 mutant
transgenic mice develop features of ALS and frontotemporal lobar degeneration.
Proc. Natl. Acad. Sci. USA 106 (44), 1880918814. http://dx.doi.org/10.1073/pnas.
0908767106.
Wils, H., Kleinberger, G., Janssens, J., Pereson, S., Joris, G., Cuijt, I., Kumar-Singh, S.,
2010. TDP-43 transgenic mice develop spastic paralysis and neuronal inclusions
characteristic of ALS and frontotemporal lobar degeneration. Proc. Natl. Acad. Sci.
USA 107 (8), 38583863. http://dx.doi.org/10.1073/pnas.0912417107.
Wils, H., Kleinberger, G., Pereson, S., Janssens, J., Capell, A., Van Dam, D., Kumar-Singh,
S., 2012. Cellular ageing, increased mortality and FTLD-TDP-associated neuro-
pathology in progranulin knockout mice. J. Pathol. 228 (1), 6776. http://dx.doi.
org/10.1002/path.4043.
Wong, P.C., Pardo, C.A., Borchelt, D.R., Lee, M.K., Copeland, N.G., Jenkins, N.A., Price,
D.L., 1995. An adverse property of a familial ALS-linked SOD1 mutation causes motor
neuron disease characterized by vacuolar degeneration of mitochondria. Neuron 14
(6), 11051116.
Zu, T., Gibbens, B., Doty, N.S., Gomes-Pereira, M., Huguet, A., Stone, M.D., Ranum, L.P.,
2011. Non-ATG-initiated translation directed by microsatellite expansions. Proc.
Natl. Acad. Sci. USA 108 (1), 260265. http://dx.doi.org/10.1073/pnas.
1013343108.
R.M. Liscic European Journal of Pharmacology xxx (xxxx) xxx–xxx
5

File (1)

ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Importance Preclinical studies have shown that neurotrophic growth factors (NTFs) extend the survival of motor neurons in amyotrophic lateral sclerosis (ALS) and that the combined delivery of these neurotrophic factors has a strong synergistic effect. We have developed a culture-based method for inducing mesenchymal stem cells (MSCs) to secrete neurotrophic factors. These MSC-NTF cells have been shown to be protective in several animal models of neurodegenerative diseases.Objective To determine the safety and possible clinical efficacy of autologous MSC-NTF cells transplantation in patients with ALS.Design, Setting, and Participants In these open-label proof-of-concept studies, patients with ALS were enrolled between June 2011 and October 2014 at the Hadassah Medical Center in Jerusalem, Israel. All patients were followed up for 3 months before transplantation and 6 months after transplantation. In the phase 1/2 part of the trial, 6 patients with early-stage ALS were injected intramuscularly (IM) and 6 patients with more advanced disease were transplanted intrathecally (IT). In the second stage, a phase 2a dose-escalating study, 14 patients with early-stage ALS received a combined IM and IT transplantation of autologous MSC-NTF cells.Interventions Patients were administered a single dose of MSC-NTF cells.Main Outcomes and Measures The primary end points of the studies were safety and tolerability of this cell therapy. Secondary end points included the effects of the treatment on various clinical parameters, such as the ALS Functional Rating Scale–Revised score and the respiratory function.Results Among the 12 patients in the phase 1/2 trial and the 14 patients in the phase 2a trial aged 20 and 75 years, the treatment was found to be safe and well tolerated over the study follow-up period. Most of the adverse effects were mild and transient, not including any treatment-related serious adverse event. The rate of progression of the forced vital capacity and of the ALS Functional Rating Scale–Revised score in the IT (or IT+IM)–treated patients was reduced (from −5.1% to −1.2%/month percentage predicted forced vital capacity, P < .04 and from −1.2 to 0.6 ALS Functional Rating Scale–Revised points/month, P = .052) during the 6 months following MSC-NTF cell transplantation vs the pretreatment period. Of these patients, 13 (87%) were defined as responders to either ALS Functional Rating Scale–Revised or forced vital capacity, having at least 25% improvement at 6 months after treatment in the slope of progression.Conclusions and Relevance The results suggest that IT and IM administration of MSC-NTF cells in patients with ALS is safe and provide indications of possible clinical benefits, to be confirmed in upcoming clinical trials.Trial Registration clinicaltrials.gov Identifiers:NCT01051882 and NCT01777646
Article
Full-text available
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are neurodegenerative disorders, related by signs of deteriorating motor and cognitive functions, and short survival. The cause is unknown and no effective treatment currently exists. For ALS, there is only a drug Riluzole and a promising substance arimoclomol. The overlap between ALS and FTD occurs at clinical, genetic, and pathological levels. The majority of ALS cases are sporadic (SALS) and a subset of patients has an inherited form of the disease, familial ALS (FALS), with a common SOD1 mutation, also present in SALS. A few of the mutant genes identified in FALS have also been found in SALS. Recently, hexanucleotide repeat expansions in C9ORF72 gene were found to comprise the largest fraction of ALS- and FTD-causing mutations known to date. TAR DNA-binding protein 43 (TDP-43), encoded by the TARDBP gene, has been identified as the pathological protein of FALS, SALS and, less frequently, FTD. The less frequent TDP-43 pathology in other forms of familial FTD has been linked to a range of mutations in GRN, FUS/TLS, rarely VCP, and other genes. TDP-43 and FUS/TLS have striking structural and functional similarities, most likely implicating altered RNA processing as a major event in ALS pathogenesis. The clinical overlap of the symptoms of FTD and ALS is complemented by overlapping neuropathology, with intracellular inclusions composed of microtubule-associated protein tau, TDP-43 and less frequently FUS, or unknown ubiquitinated proteins. Furthermore, new therapeutic approaches continue to emerge, by targeting SOD1, TDP-43 or GRN proteins. This review addresses new advances that are being made in our understanding of the molecular mechanisms of both diseases, which may eventually translate into new treatment options.
Article
Full-text available
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are two progressive, fatal neurodegenerative syndromes with considerable clinical, genetic and pathological overlap. Clinical symptoms of FTD can be seen in ALS patients and vice versa. Recent genetic discoveries conclusively link the two diseases, and several common molecular players have been identified (TDP-43, FUS, C9ORF72). The definitive etiologies of ALS and FTD are currently unknown and both disorders lack a cure. Glia, specifically astrocytes and microglia are heavily implicated in the onset and progression of neurodegeneration witnessed in ALS and FTD. In this review, we summarize the current understanding of the role of microglia and astrocytes involved in ALS and FTD, highlighting their recent implications in neuroinflammation, alterations in waste clearance involving phagocytosis and the newly described glymphatic system, and vascular abnormalities. Elucidating the precise mechanisms of how astrocytes and microglia are involved in ALS and FTD will be crucial in characterizing these two disorders and may represent more effective interventions for disease progression and treatment options in the future.
Article
Full-text available
Introduction Neurofibrillary tangles (NFT) composed of Tau are hallmarks of neurodegeneration in Alzheimer disease. Transgenic mice expressing full-length pro-aggregant human Tau (2N4R Tau-ΔK280, termed TauΔK) or its repeat domain (TauRD-ΔK280, TauRDΔK) develop a progressive Tau pathology with missorting, phosphorylation, aggregation of Tau, loss of synapses and functional deficits. Whereas TauRDΔK assembles into NFT concomitant with neuronal death, TauΔK accumulates into Tau pretangles without overt neuronal loss. Both forms cause a comparable cognitive decline (with onset at 10mo and 12mo, respectively), which is rescued upon switch-off of transgene expression. Since methylene blue (MB) is able to inhibit Tau aggregation in vitro, we investigated whether MB can prevent or rescue Tau-induced cognitive impairments in our mouse models. Both types of mice received MB orally using different preventive and therapeutic treatment protocols, initiated either before or after disease onset. The cognitive status of the mice was assessed by behavior tasks (open field, Morris water maze) to determine the most successful conditions for therapeutic intervention. Results Preventive and therapeutic MB application failed to avert or recover learning and memory deficits of TauRDΔK mice. Similarly, therapeutic MB treatment initiated after onset of cognitive impairments was ineffective in TauΔK mice. In contrast, preventive MB application starting before onset of functional deficits preserved cognition of TauΔK mice. Beside improved learning and memory, MB-treated TauΔK mice showed a strong decrease of insoluble Tau, a reduction of conformationally changed (MC1) and phosphorylated Tau species (AT180, PHF1) as well as an upregulation of protein degradation systems (autophagy and proteasome). This argues for additional pleiotropic effects of MB beyond its properties as Tau aggregation inhibitor. Conclusions Our data support the use of Tau aggregation inhibitors as potential drugs for the treatment of AD and other tauopathies and highlights the need for preventive treatment before onset of cognitive impairments. Electronic supplementary material The online version of this article (doi:10.1186/s40478-015-0204-4) contains supplementary material, which is available to authorized users.
Article
Overlap between motor neuron disease (MND) and frontotemporal dementia (FTD) occurs at clinical, genetic and pathological levels, and has been recently strengthened through the discovery of the C9orf72 genetic expansion. MND is now considered to be a multisystem disorder in which cognitive involvement may be present and, in some cases, may evolve to frank FTD. Identifying cognitive features in MND can be challenging, while, similarly, motor dysfunction in FTD may be overlooked. As such, the present review aims to decipher the variety of overlapping clinical features across the MND–FTD continuum.
Article
Behavioural/functional disturbances, characteristic of frontotemporal dementia (FTD), are also a feature of amyotrophic lateral sclerosis (ALS) and patients with combined ALS and FTD (FTD-ALS). To investigate the progression of behavioural disturbances in ALS and FTD using the FTD functional rating scale (FTDFRS). Patients with ALS, FTD-ALS and FTD were recruited from specialist clinics. Baseline assessments included the FTDFRS and the ALS functional rating scale-revised (ALSFRS-R). Baseline assessments were included, as were longitudinal assessments in a proportion of patients. In total, 21 ALS, 12 FTD-ALS and 14 behavioural variant FTD (bvFTD) patients were included in the study. Moderate or severe behavioural disturbance was common in patients with ALS at baseline (47.6%), although less frequent than in bvFTD patients; patients with FTD-ALS displayed intermediate impairment. The ALSFRS-R showed the opposite pattern and did not correlate with the FTDFRS. During the follow-up period, significant (P < 0.05) behavioural deterioration was demonstrated in patients with bvFTD and FTD-ALS, with a trend for decline in patients with ALS (P = 0.06). Motor disturbance is the primary marker of disease severity in ALS, but behavioural and functional impairment are common, and may decline independently of motor function. As such, the FTDFRS may provide valuable information in the assessment and monitoring of ALS. © 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.
Article
To estimate the incidence of early-onset dementias in a defined area of Cambridgeshire served by Addenbrooke's Hospital. The area selected for the study was that covered by the local authority areas of Cambridge City, East Cambridgeshire, and South Cambridgeshire. Cases were identified from the specialist memory and dementia clinics held at Addenbrooke's Hospital and were defined as those patients resident in the target area at the time of diagnosis, who were diagnosed with dementia before the age of 65 years between June 1, 2000, and May 31, 2006. No obvious pattern by sex was present. The incidence for all cases of primary dementia for the age range 45-64 years was estimated to be 11.5 cases per 100,000 person-years (95% CI 8.6-15.0). The incidence of frontotemporal dementia for the age range 45-64 years was estimated to be 3.5 cases per 100,000 person-years (95% CI 2.0-5.7); for Alzheimer disease the incidence for the same group was 4.2 (95% CI 2.5-6.6) and for Huntington disease the incidence rate of cases becoming affected (with or without dementia) was 0.8 (95% CI 0.2-2.3). If the incidence rates were extrapolated across England and Wales, in the region of 460 new cases of frontotemporal dementia and 550 new cases of Alzheimer disease could be expected each year in the 45-64 years age group.
Article
To establish survival in patients with pathologically confirmed frontotemporal dementia (FTD) and to determine whether clinical or pathologic subtype affects prognosis. The authors reviewed the presenting clinical features of 61 patients with dementia and pathologically confirmed FTD studied in Sydney (n = 31) and Cambridge (n = 30) over a 10-year period. Data were available on time of symptom onset, diagnosis, institutionalization, and death. Cases were classified pathologically as tau-positive and tau-negative. Of the 61 patients with FTD, 26 presented with frontal variant (fvFTD), 9 with semantic dementia, 8 with progressive nonfluent aphasia (PNFA), 9 with associated motor neuron disease (FTD-MND), and 9 with corticobasal degeneration features. There was no difference between the groups in age at symptom onset (overall mean 58.5 +/- 7.8 years), but at diagnosis the PNFA (68.3 +/- 2.7) group was significantly older than the fvFTD (59.9 +/- 7.4) and FTD-MND (57.7 +/- 7.9) groups. The median survival from symptom onset and from diagnosis was 6 +/- 1.1 years (95% CI) for fvFTD and 3 +/- 0.4 years for FTD-MND. Survival across subgroups was equivalent except for the FTD-MND group, which had significantly shorter survival. Cases with tau-positive pathology had an older age at onset and a significantly better prognosis: median survival 9.0 +/- 0.9 years vs 5.0 +/- 1.1 years. FTD is a malignant disorder with limited life expectancy. FTD-MND has the shortest duration both before and after diagnosis. Tau-positivity is associated with a more slowly progressive form of FTD.
Article
At present, there are very few therapeutic options for patients affected by amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). However, almost all patients affected by ALS or tau-negative FTD share in their brains the presence of aggregated TDP-43, a nuclear factor that plays an important role in regulating RNA metabolism. For this reason, this protein represents a very promising target to develop novel therapeutic options. Over the years, these options have mostly involved the search for new effectors capable of reducing aberrant aggregation or enhancing its clearance by UPS-dependent protein quality control or autophagy system. Targeting eventual mutations in the sequence of this protein might represent a parallel alternative therapeutic option. To this date, the study of various patient populations has allowed to find more than 50 mutations associated with disease. It is, therefore, important to better understand what the functional consequences of these mutations are. As discussed in this review, the emerging picture is that most TDP-43 mutations appear to directly relate to specific disease features such as increased aggregation, half-life, or altered cellular localization and protein-protein interactions.
Article
AMYOTROPHIC lateral sclerosis (ALS) is a degenerative disorder of motor neurons in the cortex, brainstem and spinal cord1,2. Its cause is unknown and it is uniformly fatal, typically within five years3. About 10% of cases are inherited as an autosomal dominant trait, with high penetrance after the sixth decade4,5. In most instances, sporadic and autosomal dominant familial ALS (FALS) are clinically similar4,6,7. We have previously shown that in some but not all FALS pedigrees the disease is linked to a genetic defect on chromosome 21q (refs 8, 9). Here we report tight genetic linkage between FALS and a gene that encodes a cytosolic, Cu/Zn-binding superoxide dismutase (SOD1), a homodimeric metalloenzyme that catalyzes the dismutation of the toxic superoxide anion O2.- to O2 and H2O2 (ref. 10). Given this linkage and the potential role of free radical toxicity in other neurodenegerative disorders11, we investigated SOD1 as a candidate gene in FALS. We identified 11 different SOD1 missense mutations in 13 different FALS families.