ArticlePDF AvailableLiterature Review

Current and Promising Therapies in Autosomal Recessive Ataxias

Authors:

Abstract and Figures

Background & objective: Ataxia is clinically characterized by unsteady gait and imbalance. Cerebellar disorders may arise from many causes such as metabolic diseases, stroke or genetic mutations. The genetic causes are classified by mode of inheritance and include autosomal dominant, X-linked and autosomal recessive ataxias. Many years have passed since the description of Friedreich's ataxia, the most common autosomal recessive ataxia, and mutations in many other genes have now been described. The genetic mutations mostly result in the accumulation of a toxic metabolite which causes Purkinje neuron loss and eventual cerebellar dysfunction. Unfortunately, the recessive ataxias remain a poorly known group of diseases and most of them are yet untreatable. Conclusion: The aim of this review is to provide a comprehensive clinical profile and to review the current available therapies. We overview the physiopathology, neurological features and diagnostic approach of the common recessive ataxias. The emphasis is also put on potential drugs currently or soon-to-be in clinical trials. Promising gene therapies raise the possibility of treating specifically Friedreich's ataxia, Ataxia-telangiectasia, Wilson's disease and Niemann-Pick disease in the next few years.
Content may be subject to copyright.
CNS & Neurological Disorders - Drug Targets
ISSN: 1871-5273
eISSN: 1996-3181
Impact
Factor:
2.506
SCIENCE
BENTHAM
New case of
MS Patients Untreated
patients
IFN-b treated
patients
Sampling
Flowcytometery
Send Ord ers for R eprints to reprints@benthamscience.ae
CNS & Neurological Disorders - Drug Targets, 2018, 17, 161-171
161
REVIEW ARTICLE
Current and Promising Therapies in Autosomal Recessive Ataxias
Vincent Picher-Martel1,2,* and Nicolas Dupré2,*
1Research Centre of Institut Universitaire en Santé Mentale de Québec and Department of Psychiatry and Neuroscience,
Laval University, 2601 Chemin de la Canardière, Québec, QC, G1J 2G3, Canada; 2Department of Medicine, Faculty of
Medicine, Laval University and CHU de Québec-Laval University, Axe Neurosciences, 1401, 18th Street, Québec, QC,
Canada, G1J 1Z4
A R T I C L E H I S T O R Y
Received: May 30, 2 017
Revised: December 18, 20 17
Accepted: April 01, 2018
DOI:
10.2174/1871527317666180419115029
Abstrac t: Background & Objective: Ataxia is clinically characterized by unsteady gait and imbal-
ance. Cerebellar disorders may arise from many causes such as metabolic diseases, stroke or genetic
mutations. The genetic causes are classified by mode of inheritance and include autosomal dominant,
X-linked and autosomal recessive ataxias. Many years have passed since the description of the Frie-
dreich’s ataxia, the most common autosomal recessive ataxia, and mutations in many other genes h ave
now been described. The genetic mutations mostly result in the accumulation of toxic metabolites
which causes Purkinje neuron lost and eventual cerebellar dysfunction. Unfortunately, the recessive
ataxias remain a poorly known group of diseases and most of them are yet untreatable.
Conclusion: The aim of this review is to provide a comprehensive clinical profile and to review the
currently available therapies. We overview the physiopathology, neurological features and diagnostic
approach of the common recessive ataxias. The emphasis is also made on potential drugs currently or
soon-to-be in clinical trials. For instance, p romising gene therapies raise the possibility of tr eating dif-
ferently Friedreich’s ataxia, Ataxia-t elangiectasia, Wilson’s disease and Niemann-Pick disease in the
next few years.
Keywords: Recessive ataxia, friedreich’s ataxia, ataxia-telangiectasia, ataxia with v itamin E deficiency, refsum’s disease, wil-
son’s disease, cerebrotendinous xantomatosis, niemann-pick disease type C, ataxia with oculomotor apraxia, autosomal reces-
sive spastic ataxia of charlevoix-saguenay.
1. INTRODUCTION
Symptoms of ataxia are common complaints among pa-
tients in neurology clinics. The term ataxia is generally used
to designate the neurologic conditions affecting the cerebel-
lum, which is implicated in movement coordination. Its dys-
function can cause a variety of symptoms including loss of
hand coordination and difficulty with fine motor tasks, nys-
tagmus, tremor, dysarthria, walking and balance difficulties.
Many etiologies account for this group of diseases. Although
an acute onset of cerebellar dysfunction is often caused by
infarction or hemorrhage, the differential diagnosis of
chronic degeneration is wide and contains, for instance, ge-
netic disorders, metabolic disorders, auto-immune diseases,
infections and drug-induced toxicity [1].
*Address correspondence to these authors at the Research Centre of Institut
Universitaire en Santé Mentale de Québec and Department of Psychiatry
and Neuroscience, Laval University, 2601 Chemin de la Canardière, Qué-
bec, QC, G1J 2G3, Canada; Tel: 418-455-0982;
E-mail: vince nt.picher-martel.1@ulaval.ca and Department of Medicine,
Faculty of Medicine, Laval University and CHU de Québec- Laval Univer-
sity, Axe neurosciences, 1401, 18th Street, Québec, QC, Canada, G1J 1Z4;
Tel: 418-649-0252 (63177); Fax: 418-649-5915;
E-mail: nicolas.dupre.cha@ssss.gouv.qc.ca
Hereditary conditions are frequent and numerous genes
are linked to ataxic syndromes. It is useful to classify the
hereditary ataxia by mode of inheritance. The autosomal
dominant ataxias, also referred to spinocerebellar ataxias, ar e
caused by CAG expansions into more than 35 different
genes. The clinical spectrum of sy mptoms usually appears on
average after 30 years of age and a family history of ataxic
syndrome is generally noted [2]. If the mode of inheritance
suggests an X-linked inheritance, the clinician should con-
sider the possibility of X-associated tremor-ataxia syndrome
or adrenomyeloneuropathy. On the other hand, autosomal
recessive ataxias should be considered in cases of affected
siblings and in consanguine families with healthy parents.
However, sporadic cases remain th e most common pr esenta-
tion in western countries.
Autosomal recessive cerebellar ataxias (ARCA) are a
heterogeneous group of diseases. ARCAs generally manifest
before the age of 30 with a progressive disorder of gait and
balance. The number of the identified causative gene has
grown in the past years with the advanced technologies in
genetic screening. The prevalence of all ARCAs is estimated
to be 3.3/100 000 inhabitants worldwide. However, the
prevalence varies between countries and some ARCAs are
1996-3181/18 $58.00+.00 © 2018 Bentham Science Publishers
162 CNS & Neurological Disorders - Drug Targets, 2018, Vol. 1 7, No. 3 Picher-Marte l and Dup
particularly restricted to isolated regions [3]. Compared to
other causes of cerebellar dysfunction, systemic manifesta-
tions are peculiar findings in most ARCAs and are useful to
establishing the diagnosis. Once the diagnosis of ARCA is
suspected, the clinician should perform a magnetic resonance
imaging (MRI) to determine the presence or the absence of
cerebellar atrophy and perform electromyography (EMG) to
characterize an associated neuropathy. ARCAs can be cate-
gorized according to the presence of a pure sensory neuropa-
thy, a sensorimotor neuropathy or the absence of a neuropa-
thy [4]. Finally, the diagnosis of ARCA is generally con-
firmed by gene sequencing.
The aim of this review is to highlight novel therapies for
common recessive ataxias. An emerging number of studies
have been published in the last decade. The present paper is an
updated version of a previous review written by Martineau L,
Moreau N, and Dupre N in 2014 [5]. We reviewed advances
in treatments for Friedreich’s ataxia, Ataxia-telangiectasia,
Ataxia with vitamin E deficiency, Refsum’s disease, Wilson’s
disease, Cerebrotendinous xantomatosis, Niemann-Pick dis-
ease type C, Ataxia with oculomotor apraxia and Autosomal
Recessive Spastic Ataxia of Charlevoix-S aguenay. Emphasis
is made on therapies with promising results in clinical stud-
ies and we discussed of few pre-clinical studies where the
results are highly optimistic. Although we only stated about
the medical approach, it is important to mention that patients
with ARCA certainly benefit from physiotherapy and occu-
pational therapy. Recently, a systemic review of rehabilita-
tion highlighted the effectiveness of balance exercises and
gait training [6]. The young patients also benefit from a
multidisciplinary team that includes pediatrician, neuro logist,
physiotherapist, nurses and other medical specialists.
2. AUTOSOMAL RECESSIVE CEREBELLAR ATA-
XIAS
2.1. Friedreich’s Ataxia
Friedreich’s ataxia (FRDA) was described in 1863 by
Nikolaus Friedreich and is the most common autosomal re-
cessive ataxia. Guanine-adenine-adenine (GAA) trinucleo-
tide repeat expansion in the frataxin (FXN) gene, located on
chromosome 9q13, accounts for the majority of FRDA cases
[7]. Frataxin is a mitochondrial protein implicated in iron
metabolism, oxidative stress, energy metabolism and other
mitochondrial functions [8]. Defects in the frataxin protein
result in a neurodegenerative disease affecting both the cen-
tral and the peripheral nervous systems (Fig. 1). Indeed, th e
symptoms originate from a spinocerebellar degeneration, a
cerebellar pathology and a peripheral sensory neuropathy
[7]. Most of the cases exhibit progressive limb and gait
ataxias, dysarthria and lower limb areflexia. Other features
such as dysphagia, optic atrophy, weakness, positive Babin-
ski’s sign, loss of position and vibration sense are also com-
monly found [9]. The age at onset is generally 10-12 years of
age and patients usually die between 30 to 40 years of age.
Non-neurological features such as diabetes, scoliosis and
cardiomyopathies are frequent in FRDA (Table 1). The diag-
nosis is based on clinical suspicion and genetic testing.
There is currently no therapy for Friedreich’s ataxia. Cur-
rent therapeutic concepts can be separated into three strate-
gies. The first proposed strategy is grounded on increasing
the mitochondrial function to substitute from the normal
FXN function, whereas the second and the third strategies are
based on enhancing the FXN gene expression with small
molecules or with the use of gene therapy. As a mitochon-
drial function enhancer, the antioxidant combination of vi-
tamin E with coenzyme Q10 showed some improvement in
the International Cooperative Ataxia Rating Scale (ICARS)
in two studies. The first study, a 4-years follow-up on 10
patients showed an improvement in the ICARS scores when
compared to cross-sectional data from 77 FRDA patients but
failed to demonstrated any positive effect on posture, gait
and hand dexterity [10]. The second study, a double-blind
multiple-dose study, demonstrated better than expected IC-
ARS scores in 49% of patients [11]. However, no placebo
group was used for comparison. Besides, idebenone, a struc-
tural analog of coenzyme Q10, have been widely studying in
FRDA but the results were conflicting and this compound is
not recommended. Essentially, some studies demonstrated an
improvement in cardiac structure and function but without
any improvement in neurological outcomes [12-14]. Two
open-label trials found either a significant reduction in the
ICARS scores or a stabilization of neurological symptoms in
both the pediatric and the adult populations [15, 16].
Though, these results were not reproduced in additional dou-
ble-blinded, placebo-controlled trials [17, 18].
There is evidence of iron accumulation in mitochondria
of FRDA patients but the detailed pathological mechanisms
are still unclear [19]. Therefore, the iron chelator deferiprone
have been studied in FRDA but the results were also con-
flicting and this treatment is not recommended either. Using
apparent transverse relaxation rate on MRI, a group demon-
strated a reduced iron accumulation but no significant neuro-
logical amelioration in patients treated with deferiprone [20].
Following this study, a multicentre randomized placebo-
controlled trial tested three different doses of deferiprone
(20, 40 and 60 mg/kg/day). Deterioration of ataxia symptoms
was observed in patients treated with 40 and 60 mg/kg/day
and inconclusive results were observed with the lowest dose
[19]. Besides, a combined therapy of deferiprone and idebe-
none demonstrated a slight improvement in cardiac hyper-
trophy, a progress in kinetic functions but a worsening of
gait and posture scores [21]. A triple therapy with darbepo-
etin alfa, idebenone and riboflavin showed a reduction in
disease evolution with a stability in cardiac hypertrophy dur-
ing the first two years of treatment [22] (Table 2).
Recent studies have tried to understand the effect of in-
creased frataxin protein expression levels in animal models
and in human patients. Erythropoietin was shown to have a
positive effect on frataxin expression in primary culture of
patients lymphocytes [23]. However, clinical studies failed to
demonstrate any improvement of neurological symptoms
[24-26]. Interferon-gamma (IFNγ), a cytokine implicated in
iron metabolism, increased frataxin level in cell lines and in
a mouse model of FRDA. Therefore, the sensorimotor per-
formance was strongly upgraded in these mice [27]. Follow-
ing the pre-clinical studies, an open-label trial tested subcu -
taneous injection of IFNγ during 12 weeks on 12 children
with FRDA. IFNγ was well tolerated and improved the Frie-
dreich Ataxia Rating Scale (FARS) scores [28]. A small in-
crease of the frataxin protein level was measured in the
Current and Promising Therapies in Autosomal Recessive Ataxias CNS & Neurological Disorders - Drug Targets, 2018, Vol. 17, No. 3 163
Fig. (1). Pathological mechanisms of autosomal recessive cerebellar ataxias. A) In healthy individuals, iron is transferred from cytosol to
mitochondria and iron-sulfur clusters (ISC) are formed. GAA repeats in Frataxin (FXN) gene and FXN deficiency imp airs ISC fo rmation in
the mitochondria and leads to iron responsive element binding protein 1 (IRP1) activation and causes an increased cellular iron uptake by
transferrin receptor protein 1 (TfR1). FXN deficiency also interferes with Heme synthesis. Finally, iron accumulation into mitochondria leads
to oxidative damage and Purkinje cells death. B) Ataxia-telangiectasia mutated (ATM) is implicated in coordination of reparation in DNA
double-strain breaks. Reduced ATM protein levels increase sensitivity to DNA breaks and lead to tumors, telangiectasia, ataxia and more. C)
Mutations in Phytanoyl-CoA Hydroxylase (PHYH) or peroxisomal targeting signal 2 (PTS2) genes lead to phytanic acid (PA) accumulation,
normally degraded by PhyH. PA accumulates in membranes and causes mitochondrial defects. D) Mutation in α-tocopherol transfer protein
(TTPA) gene causes a reduction of vitamin E incorporation into low density lipoproteins (LDL) in the liver. This results in reduced vitamin E
in tissue and oxidative damage in Purkinje neurons. E) Mutation in copper-transporting P-type ATPase (ATP7B) gene reduces copper clear-
ance into bile and increases copper accumulation into tissues. F) Mitochondrial sterol 27-hydroxylase is implicated in oxidation of cholesterol
into-27-hydroxycholesterol, which is subsequently needed for bile acids formation. Accumulation of cholesterol in brain can affects myelin
formation and disrupts blood-brain barrier (BBB). G) Cholesterol particles are normally trafficked from endosomal vesicles to Golgi and
endoplasmic reticulum (ER) apparatus. Niemann-Pick Type 1 and 2 (NPC1-2) mutations inhibit this trafficking and cause an accumulation in
intracellular compartments.
blood. More recently, GIFT-1, a phase IIa clinical trial, tested
the efficacy of IFNγ on 9 adult patients. Unfortunately, the
study failed to demonstrate any significant variations in the
frataxin level and in the neurological scores [29]. A phase III
clinical trial is currently ongoing.
Research in animal models proposed that nicotinamide
(vitamin B3) could increase frataxin level. It could act by
histone acetylation near the GAA repeat which leads to in-
creased wild-type frataxin expression [30]. An open-label
study from the United Kingdom evaluated escalating dose of
nicotinamide (2-8g) on 10 eligible patients. A sustained and
significant upregulation of frataxin level was observed with a
daily dose of 3 to 6 g but no significant changes on clinical
scales were observed [31].
Such as in other neurodegenerative disorders, gene ther-
apy is a promising approach in FRDA. This approach is not
yet tested in human but has demonstrated interesting results
in animal models. Interestingly, an increased expression of
FXN gene in drosophila caused a harmful effect on the fly
phenotype and suggested that level of frataxin must be care-
fully controlled in gene therapy approaches [32]. Inversely,
AAV-mediated specific cardiac expression of human frataxin
164 CNS & Neurological Disorders - Drug Targets, 2018, Vol. 1 7, No. 3 Picher-Marte l and Dup
Table 1. Clinical features of the recessive ataxias.
Recessive Ataxias Gene (Protein) Age at Onset
(Years of Age) Special Features Laboratory and Ima ging
Findings
Friedreich’s ataxia FXN (Frataxin) 10-12 Areflexia, dysphagia, optic atrophy, weakness,
Babinski sign, peripheral neuropathy, cardiomyopa-
thy, diabetes, scoliosis
No cerebellar atrophy
Spinal cord atrophy
Ataxia with vitamin E
deficiency
TTPA (α-tocopherol
transfer protein)
5-15 Loss of proprioception, areflexia, Babinski sign,
decrease visual acuity, head tremor, intellectual
decline, dystonia, retinitis pigmentosa, sensory
disturbances
Decreased serum Vit E level
No cerebellar atrophy
Spinal cord atrophy
Wilson’s disease ATP7B (copper-
transporting P-type
ATPase)
20-40 Dystonia, parkinsonism, flapping tremor, altered
cognition, seizure, personality changes, abnormal
behaviour, anxiety, depression
Reduced serum copper and ceru-
loplasmin
Elevated hepatic transaminase
Aminoaciduria
Increased copper urinary excretion
Tectal plate hyperintensity and face
of giant panda sign in MRI
Ataxia-telangiectasia ATM (ataxia
telangiectasia,
mutated)
1-2A Dysphagia, involuntary movements, abnormal eye
movement, oculocutaneous telangiectasia, cancer,
immune deficiency, pulmonary diseases, radiation
sensibility
Elevated alpha-fetoprotein
Igg deficiency
Cerebellar atrophy
Refsum’s disease PHYH (Phytanoyl-
CoA Hydroxylase)B
0-50 Infantile: early onset, hepatic and cerebral dysfunc-
tion, dysmorphia, developmental abnormalities and
infancy death
Classical: retinitis pigmentosa, anosmia, polyneuropa-
thy, deafness, cardiac arrhythmias, ichthyosis
Elevated serum phytanic acid
No cerebellar atrophy
Cerebrotendinous
xantomatosis
CYP27A1 (sterol 27-
hydroxylase)
10-50C Parkinsonism, UMN weakness, epilepsy, intellectual
disability, d ementia, psychiatric symptoms, dysto-
nia, peripheral neuropathy, cataracts, xanthomas,
hypothyroidism, cardiovascular disease, infantile
diarrhea, neonatal cholestatic jaundice
Elevated serum cholestenol
Variable cerebellar atrophy
Cerebellar of cerebral leukodystro-
phy
Niemann-Pic k Dis-
ease
NPC1/2 (intracellular
cholesterol transpor-
ter 1/2)
6-15D Movement disorder, dysphagia, supranuclear
ophtalmoplegia, cataplexy, learning and writing
difficulty, psychiatric symptoms and dementia
Variable cerebellar of brain atrophy
Ataxia with oculomo-
tor apraxia
APTX (aprataxin) 2-10 AOA1: Early and severe oculomotor apraxia,
marked dystonia, severe neuropathy, AOA2: Mild
oculomotor apraxia, low dystonia, low neuropathy,
AOA3: Severe oculomotor apraxia, severe neuropa-
thy, no dystonia, AOA4: Severe oculomotor apraxia,
marked dystonia, severe neuropa thy, cognitive
impairment
Elevated serum LDL (AOA1) low
albumin (AOA1)
Elevated serum alpha-fetoprotein
(AOA2)
Cerebellar atrophy (AOA1-2)
ARSACS ARSACS (sacsin) 1-2 Progressive spasticity, distal muscle atrophy, myeli-
nated fibers in optic disk, brisk refle x, bilateral
Babinski sign
Anterior superior cerebellar atro-
phy, variable T2 liner hypo-
densities in pons
AAtaxia appears when children begin to walk, but is highly variable between affected children; B 90% of cases are caused by mutation PHYH and 10% are caused by mutation in PTS2
Receptor (PEX7) gene; CAverage age at onset is 19 years; DJuvenile presentation is the most common; UMN upper motor neuron; ARSACS autosomal recessive spastic ataxia of
Charlevoix-Saguenay; Igg immunoglobulin.
gene in an FRDA mouse model completely prevented car-
diac disease and managed to reverse cardiomyopathy when
injected after the onset [33]. Similarly, lentiviral FXN ex-
pression in human FRDA fibroblasts improved cell viability
and ameliorated DNA damage-repair capacity [34].
2.2. Ataxia with Vitamin E Deficiency
Ataxia with vitamin E deficiency (AVED) is an infre-
quent autosomal recessive ataxia and one the rare treatable
ataxias [35]. AVED is caused by mutations in the α-
tocopherol transfer protein gene (TTPA) on chromosome
8q13.1 [36]. This protein normally supports the production
of low density lipoproteins in the liver by incorporations of
α-tocopherol (vitamin E). The resulting vitamin E deficiency
leads to Purkinje neurons defect in cerebellum [37]. Abe-
talipoproteinemia and neuroacanthocytosis syndromes, char-
acterized by mutations in the microsomal triglyceride trans-
fer protein gene (MTTP), lead to irregularity in the fat ab-
sorption and are also a source of AVED [38-40].
Current and Promising Therapies in Autosomal Recessive Ataxias CNS & Neurological Disorders - Drug Targets, 2018, Vol. 17, No. 3 165
Table 2. Treatments in recessive ataxias.
Recessive Ataxias Approved and Supportive Potential
Friedreich’s ataxia - Vitamin E with Coenzyme Q10
Inteferon gamma
Gene therapy
Ataxia with vitamin E deficiency Vitamin E -
Wilson’s disease D-penicillamine
Trientine
Zinc
Tetrathiomolybdate
Gene therapy
Ataxia-telangiectasia Physical, occupational and speech therapies,
Dopamine agonist, anticholinergics, SSRI
Betamethasone/Dexamethasone
Gene therapy
Refsum’s disease 1. Dietary restrictions
2. Plasmapheresis
3. Lipid apharesis
Liver transplantation
Cerebrotendinous xantomatosis Chenodeoxycholic acid
Ursodeoxycholic acid
Cholic acid
Taurocholic acid
-
Niemann-Pick disease Miglustat Cyclodextrin
Histone deacetylase inhibitors
Gene therapy
Ataxia with oculomotor apraxia - -
ARSACS Baclofen -
SSRI selective serotonin reuptake inhibitors; ARSACS autosomal recessive spastic ataxia o f Charlevoix-Saguenay.
AVED patients have a clinical syndrome with many fea-
tures resembling FRDA. They exhibit a progressive ataxia
with dysarthria, areflexia, cardiomyopathy and peripheral
neuropathy. However, the patients usually have retinitis
pigmentosa and head tremor which help to distinguish from
the clinical presentation of FRDA [41, 42]. Abetalipopro-
teinemia also specifically includes a vitamin A deficiency-
associated progressive retinal degeneration, acanthocytosis,
low serum triglycerides/total cholesterol and an absence of
serum beta-lipoproteins [40, 43]. Diagnosis of AVED is
based on clinical findings, low plasma level of vitamin E and
genetic testing for a mutation in TTPA [44]. Neurological
features in all forms of AVED can be improved with high
doses of vitamin E and other fat-soluble vitamins supple-
mentation [44].
2.3. Wilson’s Disease
Samuel Alexander Kinnier Wilson first described the
familial nervous system disease with cirrhosis in 1912 [45].
Wilson’s disease (WD) is characterized by reduced biliary
excretion of copper which initiates a copper accumulation in
the brain and in the liver, such as other organs. It is caused
by mutations in ATP7B gene which encode a copper-
transporting P-type ATPase [46-48]. The resulting copper
accumulation leads to mitochondrial dysfunctions with oxi-
dative stress and free radical formation [49]. An untreated
WD will occasion liver cirrhosis accompanying with a se-
vere neurologic disorder and ultimately death. WD generally
presents in young adult but is also well described in infants
and adults over 70 years of age. The typical neurological
features include dystonia, parkinsonism, dysarthria and flap-
ping tremor. Though, ataxia is rarely the primary complaint.
In addition, almost every patient with neurologic features
have Kayser-Fleischer rings and psychiatric symptoms can
be present, along with abnormal behaviours, personality
changes and cognitive impairment [49].
Wilson’s disease is one of the rare treatable recessive
ataxias but treatment must be lifelong. Treatment can be
achieved by removing the accumulated copper in the acute
phase and by preventing the subsequent accumulation with a
maintenance drug. The copper chelators D-penicillamine and
trientine are the first-line therapies for copper removing.
They act by binding copper in blood and tissues and drive its
urinary excretion. Zinc salt is an alternative treatment wh ich
inhibits the intestinal absorption of copper. It is particularly
effective to prevent subsequent tissue accumulation but has
low de-coppering capacity [49]. While the hepatic symptoms
are usually improved by copper chelators, the outcomes on
neurological symptoms are less notable. A recent study sug-
gested that only 55% of patients noticed an improvement in
166 CNS & Neurological Disorders - Drug Targets, 2018, Vol. 1 7, No. 3 Picher-Marte l and Dup
their symptoms after 4 years of treatment [50]. It is also im-
portant to note that worsening of neurological symptoms can
be reported after introduction of D-penicillamine in ap-
proximately 10% of patients [51]. Indeed, it has been sug-
gested that initial copper removing from tissues raises the
copper blood level which is toxic to the brain [52]. Another
copper chelator, tetrathiomolubdate (TTM) appeared to be a
promising alternative but is not yet approved. TTM had the
advantages of fast acting, since it can restore copper level in
few weeks instead of months, and have a superior capacity of
reducing the free blood copper [53]. Finally, in addition to
pharmacological treatment, it is also recommended to adopt
a low copper diet.
The only curative treatment for WD is liver transplanta-
tion and it is considered only for liver failure or decompen-
sated cirrhosis. However, there are not enough evidences to
support the liver transplantation for patients with predomi-
nant neurologic features [54, 55]. This lack of curative
treatment for neurologic WD opens the gate for new auspi-
cious therapies such as gene therapy or hepatocyte transplan-
tation. Several groups have tried these approaches but failed
to demonstrate any long-term positive effect on animal mod-
els [56, 57]. However, AAV has recently been used for the
expression of ATP7B gene in a WD mouse model and shown
promising results. The authors observed a long-term restora-
tion of physiological copper excretion, a decreased copper
accumulation and a reduction in liver injury in mice treated
with AAV-ATP7B [58].
2.4. Ataxia-telangiectasia
Ataxia-telangiectasia (A-T) is a multisystem disorder
caused by mutations in the ataxia-telangiectasia mutated
gene (ATM) on chromosome 11q22.3 [59]. The ATM protein
is mostly implicated in the coordination of cellular responses
to DNA double-strain breaks and in oxidative stress [60].
The clinical spectrum of symptoms is wide and highly vari-
able between patients. The first neurological symptom is
progressive ataxia beginning when the children start to walk
and they eventually reach the requirement for a wheelchair in
their teenage [61]. The patients also develop involuntary
movements (choreathetosis, dystonia and tremors), abnormal
eye movements, oculocutaneous telangiectasia, dysphagia
and immune deficiency. The patients are also at higher risk
of developing pulmonary diseases, cancers, diabetes mellitus
and are particularly sensitive to radiations [61]. The early
diagnosis can be hard to make since the disorder is rare and
the telangiectasia appears only in the later phase. However,
the typical clinical presentation associated with elevated se-
rum α-fetoprotein makes the diagnosis very probable for A-
T. The confirmed diagnosis can be made with cultured cells
or by the identification of a pathological mutation in the
ATM gene.
There is currently no therapy that modifies the progres-
sion of A-T, though it is possible to reduce some of the neu-
rological symptoms. Clinicians have observed an improve-
ment of involuntary movements with dopamine agonists and
anticholinergics. Some ataxia symptoms, such as loss of bal-
ance, may respond to the selective serotonin reuptake inhibi-
tors (SSRI) fluoxetine or to the anxiolytic buspirone [62]. A
case series proposed that betamethasone could reduce symp-
toms of ataxia [63]. To test this hypothesis, a randomized
controlled trial was conducted on 13 A-T patients from 3 to 8
years of age. A reduction of 13 (-19 to -5.5) points in the
ICARS score was observed in the betamethasone group
compared to placebo group after 31 days of treatment [64].
However, the long-term safety of betamethasone remains to
be tested. Following this trial, a single-arm, open-label Phase
II trial, was conducted with a new delivery method [65]. The
encapsulation of dexamethasone into autologous erythro-
cytes allowed a one month sustained low infusion and re-
duced the side effects normally seen with long-term corticos-
teroids. After 6 months of monthly injection on 22 A-T pa-
tients, the authors observed a reduction of 4 points (-7.1 to -
0.9) in the ICARS scores. These studies suggest that corticos-
teroids may be beneficial for A-T patients but more works
remain to be performed to clarify the treatment posology.
A recen t study proposed that the beneficial effects of
corticosteroids may come from their antioxidant properties
[66]. Indeed, the antioxidant therapy approach with different
drugs has been tested in many pre-clinical studies in the last
decade [67-69]. Lavin MF et al. previously reviewed ongo-
ing clinical trials on antioxidan t or combined therapies [62].
Unfortunately, none of these trials are available in the cur-
rent literature, suggesting inconclusive or negative results on
neurological outcomes. Finally, gene therapy approaches,
using herpes simplex virus and AAV, have demonstrated
interesting results in animal models but have not yet been
tested in humans [70, 71].
2.5. Refsum’s Disease
First described in 1946, Refsum’s disease (RD) is caused
by mutations in either Phytanoyl-CoA Hydroxylase (PHYH),
also called PAHX, or PTS2 Receptor (PEX7) genes [72, 73].
It is characterized by the accumulation of phytanic acid (PA)
which is normally degraded by the phytanoyl-CoA hydroxy-
lase. The pathogenesis is not well understood but a mem-
brane distortion caused by the excessive incorporation of PA
has been suggested [74]. We can classify RD into classical,
adult or infantile forms. The infantile RD is distinguishable
by the early onset, hepatic and cerebral dysfunction with
dysmorphia, developmental abnormalities and death in in-
fancy. The classical form of RD starts in teenage and the
patients exhibit retinitis pigmentosa, anosmia, polyneuropa-
thy, deafness, ataxia, cardiac arrhythmias and ichthyosis. The
diagnosis requires elevated plasma level of PA over 200
μmol/L with either genetic testing or enzyme analysis. A n
increased level of cerebrospinal fluid proteins can also sup-
port the diagnosis [75].
The handling of RD is separated into the chronic mainte-
nance and into the treatment of the acute presentation. The
maintenance is based on dietary restrictions of PA-containing
meals. PA is mostly present in dairy products, animal fats and
green vegetables. The average consumption of PA is esti-
mated to be around 50-100 mg/day and the treatment aim is
to reduce the intake to less than 10 mg per day. A retrospec-
tive study on 13 patients succeeds to demonstrate a reduction
of 89±11% of plasma PA with dietary restriction only [76].
The reduction of plasmatic PA level was generally sufficient
to stop the progression of neurological symptoms but did not
entirely resolve them [75]. To assure the appropriate intake
Current and Promising Therapies in Autosomal Recessive Ataxias CNS & Neurological Disorders - Drug Targets, 2018, Vol. 17, No. 3 167
of micronutrients with the low-PA diet, the fat-soluble vita-
mins, vitamin B12, copper and selenium have to be periodi-
cally monitored. However, no regular supplementation is
recommended [77]. In addition to low-PA diet, patients are
advised to avoid rapid weight loss and fasting which can
stimulate lipolysis and increase PA level. The acute presenta-
tion can be controlled by plasmapheresis or lipid apheresis.
A case series of 4 patients has also suggested that lipid
apheresis can be a long-term treatment since this method is
well tolerated and efficient [78]. Liver transplantation has
been tried for infantile Refsum’s Disease and has managed to
significantly improve the neurological outcomes of these
young patients [79, 80].
2.6. Cerebrotendinous Xantomatosis
Cerebrotendinous xantomatosis (CTX) is a rare disorder
of bile metabolism caused by mutations in CYP27A1 gene on
chromosome 2q33. This gene encodes for the mitochondrial
sterol 27-hydroxylase implicated in the oxidation of choles-
terol into 27-hydroxycholesterol, which is subsequently
needed for bile acid formation [81]. The enzyme dysfunction
leads to an accumulation of cholesterol and cholestenol in
almost all tissues. The mechanisms underlying neurodegen-
eration are not well described but it is suggested that accu-
mulation of cholestenol in the brain can affect myelin forma-
tion or disrupt the blood-brain barrier [82]. The mean age at
onset is 19 years of age (10-50) but the delay in diagnosis
can easily reach 20 years [83]. Neurological features are of-
ten the initial symptoms and include ataxia, parkinsonism,
upper motor neuron weakness, epilepsy, intellectual disabil-
ity, dementia, psychiatric symptoms, dystonia and peripheral
neuropathy. The systemic non-neurological symptoms are
composed of premature cataracts, xanthomas, hypothyroid-
ism, atherosclerosis and cardiovascular disease. Infantile
diarrhea and neonatal cholestatic jaundice precede the onset
of symptoms by many years [84]. Although the confirmed
diagnosis is made with genetic analysis, typical neuroimag-
ing and laboratory findings strongly suggest the diagnosis.
An elevated blood cholestanol concentration and an in-
creased bile alcohol concentration in urine and plasma also
suggest the diagnosis. Magnetic resonance imaging can re-
veal cerebellar atrophy and bilateral cerebellar lesions in
dentate nuclei [84].
Rep lacement therapy is the treatment of choice for CTX.
A supplementation of bile acids is thought to reduced chole-
stanol concentration by activating the bile acid negative
feedback. Chenodeoxycholic acid (CDCA), ursodeoxycholic
acid (UDCA), cholic acid and taurocholic acid are currently
available for treatment of CTX [85, 86]. An important reduc-
tion in neurological symptoms and an improvement of elec-
troencephalograms and imaging was observed in a clinical
trial on 17 patients treated with CDCA [87]. CDCA is the
therapy of choice to reduced neurological symptoms whereas
cholic acid is particu larly efficient for non-neurological
symptoms. CDCA treatment must be started during the early
phase for efficient prevention of neurological deterioration
[88]. The recommended posology is 750 mg/day divided into
three doses and 15 mg/kg per day for children. However, a
recent case report suggested that 15mg/kg was hepatoxic in
infants and a maximum of 5 mg/kg/day was non-toxic and
efficient [89].
2.7. Niemann-Pick Disease Type C
Niemann-Pick disease (NP) is a group of lipid storage
disorders grounded on the description of Albert Niemman
and Ludwig Pick in the 1920s. NP type A and B are charac-
terized by a defect in sphingomyelinase acid production
while a defect in intracellular lipid trafficking is involved in
the pathogenesis of NP type C (NP-C) and leads to the ac-
cumulation of cholesterol and glycosphingolipids in tissues
[90]. NP-C is caused by mutations in NPC1 (90%) or NPC2
(4%) [91, 92] genes, both encoding for intracellular choles-
terol transporters which function together. The age at onset
can vary from the prenatal period to the late adult age and
the patients with early manifestations are normally dying
between 10 and 25 years of age [90].
The symptoms of NP-C are dependant on the age at on-
set. In prenatal and neonatal periods, the features include
hydrops fetalis, fetal ascites, jaundice and cholestatic hepa-
topathy. The infants also develop hepatosplenomegaly as a
major manifestation. However, the most common form of
NP-C begins between 6 and 15 years of age and is mostly
characterized by neurological manifestations. An ataxic syn-
drome accompanied by movement disorders, dysphagia, ver-
tical supranuclear ophthalmoplegia and cataplexy is typical
of the juvenile presentation. While the young also develop
behavioral problems accompanied by learning and writing
difficulty, the adults can develop psychiatric symptoms and
dementia. A suspicion index (SI) has been created to help the
diagnosis of NP-C [93]. The index attributes determined
points to each NP-C manifestation and separates the diagno-
sis between high, moderate and low probability. A high sus-
picion index should be referred for genetic analysis.
Miglustat, an inhibitor of glucosylceramide synthase, the
enzyme that catalyzes glycosphingolipids synthesis, is th e
only approved treatment with recognised efficacy in reduc-
ing neurological symptoms [94]. A randomized-controlled
12-months duration trial of 29 patients showed an improve-
ment in horizontal saccadic eye movement velocity, ambula-
tion, cognition and dysphagia [95]. A long-term extension
trial and a retrospective observational study also confirmed
these findings and suggested that disease progression can be
delayed with miglustat [96, 97]. The treatment regimen for
adults (≥12 years of age) is 200 mg three times a day and the
dose is calculated per body surface area under the age of 12
[95].
Cyclodextrin, a cholesterol chelator, has been shown to
be effective in mice models [98, 99]. Two patients were
treated with cyclodextrin in an initial clinical study [100].
The drug was partially effective in reducing hepatosple-
nomegaly but no effect on neurological outcomes was noted,
probably because of its incapacity to cross the blood-brain
barrier. To overcome this difficulty, the authors have per-
formed intrathecal injection of cyclodextrin in one patient
with the use of an Ommaya reservoir. The neurological
symptoms remained stable over a two years period [101].
Finally, the quest for a curative treatment is still ongoing
and promising therapies are drawing attention into the litera-
ture. Histone deacetylase inhibitors have shown some effec-
tiveness in reducing cholesterol storage in fibroblasts and in
reducing neurological symptoms in mouse models [102-
168 CNS & Neurological Disorders - Drug Targets, 2018, Vol. 1 7, No. 3 Picher-Marte l and Dup
105]. A phase III clinical trial is currently ongoing but the
results ar e not yet available. Recently, intracardiac injection
of AAV9-NPC1 gene in mice resulted in longer Purkinje cell
survival, increased lifespan and improved locomotor activity
[106].
2.8. Ataxias with Oculomotor Apraxia
Ataxias with oculomotor apraxia are a group of 4 reces-
sive ataxias with associated gene mutations: type 1 (AOA1)
is caused by mutations in APTX, type 2 (AOA2) by muta-
tions in SETX, type 3 (AOA3) by mutations in PIK3R5 and
type 4 (AOA4) by mutations in PNKP. All four types can be
differentiated by age at onset and variable degrees of symp-
toms. AOA1, the most severe AOA and the second most
common ARCA in Japan and Portugal, is characterized by
the onset of cerebellar ataxia and severe oculomotor aprax ia
at 4 (2-10) years of age. Patients with oculomotor apraxia
have difficulty with object fixation and have limited and
slow gaze. The other neurological findings include an axonal
peripheral neuropathy, as well as areflexia, dystonia, chorea
and cognitive deficits. Peculiar findings for each AOA are
shown in Table 1. The diagnosis is suggested by cerebellar
atrophy in cerebral imaging, axonal neuropathy on EMG and
genetic analysis. The treatment is mainly symptomatic and a
wheelchair is often necessary by 15-20 years of age.
2.9. Autosomal Recessive Spastic Ataxia of Charlevoix-
Saguenay
First described in the 70’ in the Charlevoix-Saguenay
region of Quebec, the autosomal recessive spastic ataxia of
Charlevoix-Saguenay (ARSACS) is now described world-
wide [107, 108]. It is caused by mutations in the ARSACS
gene on chromosome 13q12.12 which encodes the sacsin
protein [109]. The exact role of sacsin is not well described
but it seems to have a chaperone activity together with
Hsp70, which is implicated in aggregates clearing in many
neurodegenerative diseases [110]. Progressive spasticity is
the initial symptom observed in infants with ARSACS. They
also developed dysarthria, cerebellar ataxia with distal mus-
cle atrophy and nystagmus. Two findings can help distin-
guish ARSACS from the other recessive ataxias. Abnormal
myelination process may cause myelinated fibers radiating
from the optic disk on fundoscopy examination. It is also
possible to observe brisk reflexes in the middle stage of the
disease as compared to most recessive ataxias where the re-
flexes are usually decreased. Babinski’s sign is present in
almost of all cases. The diagnosis is suggested by the combi-
nation of a typical neurological examination with MRI imag-
ing which demonstrates atrophy mostly in the cerebellar
vermis. Abnormal fundoscopy and signs of a severe axonal
neuropathy confirmed on nerve conduction studies are elec-
tromyography can also improve diagnostic accuracy [108].
The diagnosis is confirmed with genetic analysis. Unfortu-
nately, there is no effective treatment for ARSACS. Baclofen
can be useful to reduce spasticity and prevent joint contrac-
tures in the early phases of the disease.
CONCLUSION
The differential diagnosis of ARCA is not easy to per-
form with clinical findings since the peculiar features are not
always present in early phases of the diseases. The accessi-
bility of the gen etic tests has completely changed the clinical
approach. It is now easier to perform the precise diagnosis
and includ e patients into the appropriate clinical trials. Hope-
fully, this will increase the number of patients in clinical
trials and these will eventually result in potential therapies.
Gene therapy approach is one of the potential treatments in
many ARCAs since most of them result from a lost-of-
function mechanism and replacement of that mutated protein
could correct the metabolic dysfunction. This approach has
been clinically tested in other neurological diseases such as
amyotrophic lateral sclerosis and gave promising results with
advanced delivering technologies [111]. Successfully, this
approach recently led to the first medical therapy for spinal
muscular atrophy [112]. Although few recessive ataxias are
treatable, more research must be performed for most ARCAs
if we are to improve patient care over the next decade.
LIST OF ABBREVIATIONS
AAV = Adeno-associated Viral
AOA = Ataxia with Oculomotor Apraxia
ARCA = Autosomal Recessive Cerebellar Ataxias
ARSACS = Autosomal Recessive Spastic Ataxia of Char-
levoix-Saguenay
A-T = Ataxia-telangiectasia
ATM = Ataxia-telangiectasia Mutated
AVED = Ataxia with Vitamin E Deficiency
CDCA = Chenodeoxycholic Acid
CTX = Cerebrotendinous Xantomatosis
EMG = Electromyography
FARS = Friedreich Ataxia Rating Scale
FRDA = Friedreich’s Ataxia
FXN = Frataxin
GAA = Guanine-adenine-adenine
ICARS = International Cooperative Ataxia Rating Scale
IFNγ = Inteferon Gamma
MRI = Magnetic Resonance Imaging
MTTP = Microsomal Triglyceride Transfer Protein
NP = Niemann-Pick Disease
NP-C = Niemann-Pick Disease Type C
PA = Phytanic Acid
PHYH = Phytanoyl-CoA Hydroxylase (PAHX)
PTS2 = Peroxisomal Targeting Signal Receptor
(PEX7)
RD = Refsum’s Disease
SSRI = Selective Serotonin Reuptake Inhibitors
TTM = Tetrathiomolubdate
TTPA = α-tocopherol Transfer Protein
UDCA = Ursodeoxycholic Acid
WD = Wilson’s Disease
Current and Promising Therapies in Autosomal Recessive Ataxias CNS & Neurological Disorders - Drug Targets, 2018, Vol. 17, No. 3 169
CONSENT FOR PUBLICATION
Not applicable.
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or
otherwise.
ACKNOWLEDGEMENTS
V.P-M. MD is a neurology resident and PhD student who
holds a Frederick Banting and Charles Best doctoral scholar-
ship from Canadian Institutes of Health Research (CIHR).
N.D. MD-MSc. is funded by CIHR and by Canadian Consor-
tium on Neurodegeneration in Aging (CCNA). V.P-M. per-
formed the review of the literature, wrote the paper and pre-
pared the tables and the figure. N.D. reviewed the paper and
supervised the project.
REFERENCES
[1] Winchester S, Singh PK, Mikati MA. Ataxia. Handb Clin Neurol
2013; 112: 1213-7.
[2] Jayadev S, Bird TD. Hereditary ataxias: Overview. Genet Med
2013; 15(9): 673-83.
[3] Ruano L, Melo C, Silva MC, Coutinho P. The global epidemiology
of hereditary ataxia and spastic paraplegia: A systematic review of
prevalence studies. Neuroepidemiology 2014; 42(3): 174-83.
[4] Anheim M, Tranchant C, Koenig M. The autosomal recessive
cerebellar ataxias. N Engl J Med 2012; 366(7): 636-46.
[5] Martineau L, Noreau A, Dupre N. Therapies for ataxias. Curr Treat
Options Neurol 2014; 16(7): 300.
[6] Marquer A, Barbieri G, Perennou D. The assessment and treatment
of postural disorders in cerebellar ataxia: A systematic review. Ann
Phys Rehabil Med 2014; 57(2): 67-78.
[7] Abrahao A, Pedroso JL, Braga-Neto P, et al. Milestones in Frie-
dreich ataxia: More than a century and still learning. Neurogenetics
2015; 16(3): 151-60.
[8] Pandolfo M, Pastore A. The pathogenesis of Friedreich ataxia and
the structure and function of frataxin. J Neurol 2009; 256(Suppl 1):
9-17.
[9] Parkinso n MH, Boesch S, Nachbauer W, Mario tti C, Giunti P.
Clinical features of Friedreich's ataxia: Classical and atypical phe-
notypes. J Neurochem 2013; 126 (Suppl 1): 103-17.
[10] Hart PE, Lodi R, Rajagopalan B, et al. Antioxidant treatment of
patients with Friedreich ataxia: four-year follow-up. Arch Neurol
2005; 62(4): 621-6.
[11] Cooper JM, Korlipara LV, Hart PE, Bradley JL, Schapira AH.
Coenzyme Q10 and vitamin E deficiency in Friedreich's ataxia:
Predictor of efficacy of vitamin E and coenzyme Q10 therapy. Eur
J Neurol 2008; 15(12): 1371-9.
[12] Mariotti C, Solari A, Torta D, et al. Idebenone treatment in Frie-
dreich patients: One-year-long randomized placebo-controlled trial.
Neurology 2003; 60(10): 1676-9.
[13] Hausse AO, Aggoun Y, Bonnet D, et al. Idebenone and reduced
cardiac hypertrophy in Friedreich's ataxia. Heart 2002 ; 87(4): 346-9.
[14] Buyse G, Mertens L, Di Salvo G, et al. Idebenone treatment in
Friedreich's ataxia: Neurological, cardiac, and biochemical moni-
toring. Neurology 2003; 60(10): 1679-81.
[15] Artuch R, Aracil A, Mas A, et al. Friedreich's ataxia: Idebenone
treatment in early stage patien ts. Neuropediatrics 2002; 33(4): 190-3 .
[16] Pineda M, Arpa J, Montero R, et al. Idebenone treatment in paedi-
atric and adult patients with Friedreich ataxia: Long-term follow-
up. Eur J Paediatr Neurol 2008; 12(6): 470-5.
[17] Di Prospero NA, Baker A, Jeffries N, Fischbeck KH. Neurological
effects of high-dose idebenone in patients with Friedreich's ataxia:
A randomised, placebo-controlled trial. Lancet Neurol 2007; 6(10):
878-86.
[18] Lynch DR, Perlman SL, Meier T. A phase 3, double-blind, pla-
cebo-controlled trial of idebenone in friedreich ataxia. Arch Neurol
2010; 67(8): 941-7.
[19] Pandolfo M, Hausmann L. Deferiprone for the treatment of Frie-
dreich's ataxia. J Neurochem 2013; 126(Suppl 1): 142-6.
[20] Boddaert N, Le Quan Sang KH, Rotig A, et al. Selective iro n
chelation in Friedreich ataxia: Biologic and clinical implications.
Blood 2007; 110(1): 401-8.
[21] Velasco-Sanchez D, Aracil A, Montero R, et al. Combined therapy
with idebenone and deferiprone in patients with Friedreich's ataxia.
Cerebellum 2011; 10(1): 1-8.
[22] Arpa J, Sanz-Gallego I, Rodriguez-de-Rivera FJ, et al. Triple ther-
apy with darbepoetin alfa, idebenone, and riboflavin in Friedreich's
ataxia: An open-label trial. Cerebellum 2013; 12(5): 713-20.
[23] Sturm B, Stupphann D, Kaun C, et al. Recombinant human
erythropoietin: Effects on frataxin expression in vitro. Eur J Clin
Invest 2005; 35(11): 711-7.
[24] Sacca F, Piro R, De Michele G, et al. Epoetin alfa increases fra-
taxin production in Friedreich's ataxia without affecting hematocrit.
Mov Disord 2011; 26(4): 739-42.
[25] Mariotti C, Fancellu R, Caldarazzo S, et al. Erythropoietin in Frie-
dreich ataxia: No effect on frataxin in a randomized controlled trial.
Mov Disord 2012; 27(3): 446-9.
[26] Boesch S, Nachbauer W, Mariotti C, et al. Safety and tolerability
of carbamylated erythropoietin in Friedreich's ataxia. Mov Disord
2014; 29(7): 935-9.
[27] Tomassini B, Arcuri G, Fortuni S et al. Interferon gamma upregu-
lates frataxin and corrects the functional deficits in a Friedreich
ataxia model. Hum Mol Genet 2012; 21(13): 2855-61.
[28] Seyer L, Greeley N, Foerster D, et al. Open-label pilot study of
interferon gamma-1b in Friedreich ataxia. Acta Neurol Scand 2015;
132(1): 7-15.
[29] Marcotulli C, Fortuni S, Arcuri G, et al. GIFT-1, a phase IIa clini-
cal trial to test the safety and efficacy of IFNgamma administration
in FRDA patients. Neurol Sci. 2016; 37(3): 361-4.
[30] Chan PK, Torres R, Yandim C, et al. Heterochromatinization in-
duced by GAA-repeat hyperexpansion in Friedreich's ataxia can be
reduced upon HDAC inhibition by vitamin B3. Hum Mol Genet.
2013; 22(13): 2662-75.
[31] Libri V, Yandim C, Athanasopoulos S, et al. Epigenetic and neuro-
logical effects and safety of high-dose nicotinamide in patients with
Friedreich's ataxia: An exploratory, open-label, dose-escalation
study. Lancet 2014; 384(9942): 504-13.
[32] Navarro JA, Llorens JV, Soriano S, et al. Overexpression of human
and fly frataxins in Drosophila provokes deleterious effects at bio-
chemical, physiological and developmental levels. PLoS One 2011;
6(7): e21017.
[33] Perdomini M, Belbellaa B, Monassier L, et al. Prevention and
reversal of severe mitochondrial cardiomyopathy by gene therapy
in a mouse model of Friedreich's ataxia. Nat Med 2014; 20(5):
542-7.
[34] Khonsari H, Schneider M, Al-Mahdawi S, et al. Lentivirus-
meditated frataxin gene delivery reverses genome instability in
Friedreich ataxia patient and mouse model fibroblasts. Gene Ther
2016; 23(12): 846-56.
[35] Pearson TS. More Than Ataxia: Hyperkinetic Movement Disorders
in Childhood Autosomal Recessive Ataxia Syndromes. Tremor
Other Hyperkinet Mov (N Y) 2016; 6: 368.
[36] Ouahchi K, Arita M, Kayden H, et al. Ataxia with isolated vitamin
E deficiency is caused by mutations in the alpha-tocopherol trans-
fer protein. Nat Genet 1995; 9(2): 141-5.
[37] Ulatowski L, Parker R, Warrier G, et al. Vitamin E is essential for
Purkinje neuron integrity. Neuroscience 2014; 260: 120-9.
[38] Sokol RJ, Guggenheim MA, Iannaccone ST, et al. Improved neu-
rologic function after long-term correction of vitamin E deficiency
in children with chronic cholestasis. N Engl J Med 1985; 313(25):
1580-6.
[39] Khatun I, Walsh MT, Hussain MM. Loss of both phospholipid and
triglyceride transfer activ ities of microsomal triglyceride transfer
protein in abetalipoproteinemia. J Lipid Res 2013; 54(6): 1541-9.
[40] Hammer MB, El Euch-Fayache G, Nehdi H, et al. Clinical features
and molecu lar genetics of two Tunisian families with abetalipopro-
teinemia. J Clin Neurosci 2014; 21(2): 311-5.
[41] Mariotti C, Gellera C, Rimoldi M, et al. Ataxia with iso lated vita-
min E deficiency: Neurological phenotype, clinical follow-up and
novel mutations in TTPA gene in Italian families. Neurol Sci 2004;
25(3): 130-7.
170 CNS & Neurological Disorders - Drug Targets, 2018, Vol. 1 7, No. 3 Picher-Marte l and Dup
[42] Di Donato I, Bian chi S, Federico A. Ataxia with vitamin E defi-
ciency: Update of molecular diagnosis. Neurol Sci 2010; 31(4):
511-5.
[43] Lee J, Hegele RA. Abetalipoproteinemia and homozygous hypobe-
talipoproteinemia: A framework for diagnosis and management. J
Inherit Metab Dis 2014; 37(3): 333-9.
[44] Schuelke M. Ataxia with Vitamin E Deficiency. In: Pagon RA,
Adam MP, Ardinger HH, Wallace SE, Amemiya A, Bean LJH, et
al., editors. GeneReviews(R). Seattle (WA): Univ ersity of Wash-
ington, Sea ttleUniversity of Washington, Seattle. GeneReviews is a
registered trademark of the University of Washington, Seattle. All
rights reserved.; 1993.
[45] Wilson SAK. Progressive lenticular degeneration: A familial nerv-
ous disease associated with cirrhosis of the liver. Brain 1912;
34(4): 295-507.
[46] Bull PC, Thomas GR, Rommens JM, Forbes JR, Cox DW. The
Wilson disease gene is a putative copper transporting P-type AT-
Pase similar to the Menkes gene. Nat Genet 1993; 5(4): 327-37.
[47] Tanzi RE, Petrukhin K, Chernov I, et al. The Wilson disease gene
is a copper transporting ATPase with homology to the Menkes dis-
ease gene. Nat Genet 1993; 5(4): 344-50.
[48] Petrukhin K, Fischer SG, Pirastu M, et al. Mapping, cloning and
genetic characterization of the region containing the Wilson disease
gene. Nat Genet. 1993; 5(4): 338-43.
[49] Bandmann O, Weiss KH, Kaler SG. Wilson's disease and other
neurological copper disorders. Lancet Neurol 2015; 14(1): 103-13.
[50] Weiss KH, Thurik F, Gotthardt DN, et al. Efficacy and safety of
oral chelators in treatment of patients with Wilson disease. Clin
Gastroenterol Hepatol 2013; 11(8): 1028-35.e1-2.
[51] EASL Clinical Practice Guidelines: Wilson's disease. J Hepatol
2012; 56(3): 671-85.
[52] Brewer GJ, Terry CA, Aisen AM, Hill GM. Worsening of neu-
rologic sy ndrome in patien ts with Wilson's d isease with initial
penicillamine therapy. Arch Neurol 1987; 44(5): 490-3.
[53] Brewer GJ, Askari F, Dick RB, et al. Treatment of Wilson's disease
with tetrathiomolybdate: V. Control of free copper by tetrathiomo-
lybdate and a comparison with trientine. Transl Res 2009; 154(2):
70-7.
[54] Medici V, Mirante VG, Fassati LR, et al. Liver transplantation for
Wilson's disease: The burden of neurological and psychiatric disor-
ders. Liver Transpl 2005; 11(9): 1056-63.
[55] Weiss KH, Schafer M, Gotthardt DN, et al. Outcome and develop-
ment of symptoms after orthotopic liver transplantation for Wilson
disease. Clin Transplant 2013; 27(6): 914-22.
[56] Malhi H, Irani AN, Volenberg I, Schilsky ML, Gupta S. Early cell
transplantation in LEC rats modeling Wilson's d isease eliminates
hepatic copper with reversal of liver disease. Gastroenterology
2002; 122(2): 438-47.
[57] Ha-Hao D, Merle U, Hofmann C, et al. Chances and shortcomins
of adenovirus-mediated ATP7B gen e transfer in Wilson disease:
Proof of principle demonstrated in a pilot study with LEC rats. Z
Gastroenterol 2002; 40(4): 209-16.
[58] Murillo O, Luqui DM, Gazquez C, et al. Long-term metabolic
correction of Wilson's disease in a murine model by gene therapy. J
Hepatol 2016; 64(2): 419-26.
[59] Gatti RA, Berkel I, Boder E, et al. Localization of an ataxia-
telangiectasia gene to chromosome 11q22-23. Nature 1988;
336(6199): 577-80.
[60] Shiloh Y, Ziv Y. The ATM protein kinase: Regulating the cellular
response to genotoxic stress, and more. Nat Rev Mol Cell Biol
2013; 14(4): 197-210.
[61] Rothblum-Oviatt C, Wright J, Lefton-Greif MA, et al. Ataxia te-
langiectasia: A review. Orphanet J Rare Dis 2016; 11(1): 159.
[62] Lavin MF, Gueven N, Bottle S, Gatti RA . Current and potential
therapeutic strategies for the treatment of ataxia-telangiectasia. Br
Med Bull 2007; 81-82: 129-47.
[63] Broccoletti T, Del G iudice E, Cirillo E, et al. Efficacy of very-low-
dose betamethasone on neurological symptoms in ataxia-
telangiectasia. Eur J Neurol 2011; 18(4): 564-70.
[64] Zannolli R, Buoni S, Betti G, et al. A randomized trial of oral be-
tamethaso ne to reduce ataxia symptoms in ataxia telang iectasia.
Mov Disord 2012; 27(10): 1312-6.
[65] Chessa L, Leuzzi V, Plebani A, et al. Intra-erythrocyte infusion of
dexamethasone reduces neurological symptoms in ataxia
teleangiectasia patients: Results of a phase 2 trial. Orphanet J Rare
Dis 2014; 9: 5.
[66] Biagiotti S, Menotta M, Orazi S, et al. Dexamethasone improves
redox state in ataxia telangiectasia cells by promoting an NRF2-
mediated antioxidant response. Febs j 2016; 283(21): 3962-78.
[67] D'Souza AD, Parish IA, Krause DS, Kaech SM, Shadel GS. Reduc-
ing mitochondrial ROS improves disease-related pathology in a
mouse model of ataxia-telangiectasia. Mol Ther 2013; 21(1): 42-8.
[68] Gueven N, Luff J, Peng C, et al. Dramatic extension o f tumor la-
tency and correction of neurobehavioral phenotype in Atm-mutant
mice with a nitroxide antioxidant. Free Radic Biol Med 2006;
41(6): 992-1000.
[69] Reliene R, Schiestl RH. Experimental antioxidant therapy in ataxia
telangiectasia. Clin Med Oncol 2008; 2: 431-6.
[70] Qi J, Shackelford R, Manuszak R, et al. Functional expression of
ATM gene carried by HSV amplicon vector in vitro and in vivo.
Gene Ther 2004; 11(1): 25-33.
[71] Cortes ML, Oehmig A, Saydam O, et al. Targeted integration o f
functional human ATM cDNA into genome mediated by
HSV/AAV hybrid amplicon vector. Mol Ther 2008; 16(1): 81-8.
[72] Mihalik SJ, Morrell JC, Kim D, et al. Identification of PAHX, a
Refsum disease gene. Nat Genet 1997; 17(2): 185-9.
[73] Van den Brink DM, Brites P, Haasjes J, et al. Identification of
PEX7 as the second gene involved in Refsum disease. Am J Hum
Genet 2003; 72(2): 471-7.
[74] Jansen GA, Waterham HR, Wanders RJ. Molecular basis of Ref-
sum disease: Sequence variations in phytanoyl-CoA hydroxylase
(PHYH) and the PTS2 receptor (PEX7). Hum Mutat 2004; 23(3):
209-18.
[75] Wanders RJA, Waterham HR, Leroy BP. Refsum Disease. In:
Pagon RA, Adam MP, Ardinger HH, Wallace SE, Amemiya A,
Bean LJH, et al., editors. Gene Reviews (R). Seattle (WA): Univer-
sity of Washington, Seattle University of Washington, Seattle. All
rights reserved.; 1993.
[76] Baldwin EJ, Gibberd FB, Harley C, et al. The effectiveness of
long-term dietary therapy in the treatment of adult Refsum disease.
J Neurol Neurosurg Psychiatry 2010; 81(9): 954-7.
[77] Baldwin EJ, Harrington DJ, Sampson B, Feher MD, Wierzbicki
AS. Safety of long-term restrictive diets for peroxisomal disorders:
Vitamin and trace element status of patients treated for Adult Ref-
sum Disease. Int J Clin Pract 2016; 70(3): 229-35.
[78] Zolotov D, Wagner S, Kalb K, et al. Long-term strategies for the
treatment of Refsum's disease using therapeutic apheresis. J Clin
Apher 2012; 27(2): 99-105.
[79] Van Maldergem L, Moser AB, Vincent MF, et al. Orthotopic liver
transplantation from a living-related donor in an infant with a per-
oxisome biogenesis defect of the infa ntile Refsum disease type. J
Inherit Metab Dis 2005; 28(4): 593-600.
[80] Matsunami M, Shimozawa N, Fukuda A, et al. Living-donor liver
transplantation from a h eterozygous parent for infantile refsum dis-
ease. Pediatrics 2016; 137(6).
[81] Cali JJ, Hsieh CL, Francke U, Ru ssell DW. Mutatio ns in the bile
acid biosynthetic enzyme sterol 27-hydroxylase underlie cerebro-
tendinous xanthomatosis. J Biol Chem 1991; 266(12): 7779-83.
[82] Salen G, Berginer V, Shore V, et al. Increased concentrations of
cholestanol and apolipoprotein B in the cerebrospinal fluid of pa-
tients with cerebrotendinous xanthomatosis. Effect of chenodeoxy-
cholic acid. N Engl J Med 1987; 316(20): 1233-8.
[83] Pilo-de-la-Fuente B, Jimenez-Escrig A, Lorenzo JR, et al. Cerebro-
tendinous xanthomatosis in Spain: Clinical, prognostic, and genetic
survey. Eur J Neurol 2011; 18(10): 1203-11.
[84] Nie S, Chen G, Cao X, Zhang Y. Cerebrotendinous xanthomatosis:
A comprehensive review of pathogenesis, clinical manifestations,
diagnosis, and management. Orphanet J Rare Dis 2014; 9: 179.
[85] Pierre G, Setchell K, Blyth J, Preece MA, Chakrapani A, McKier-
nan P. Prospective treatment of cerebrotendinous xanthomatosis
with cholic acid therapy. J Inherit Metab Dis 2008; 31(Suppl 2):
S241-5.
[86] Setchell KD, Heubi JE. Defects in bile acid biosynthesis--diagnosis
and treatment. J Pediatr Gastroenterol Nutr 2006; 43(Suppl 1): S17-
22.
[87] Berginer VM, Salen G, Shefer S. Long-term treatment of cerebro-
tendinous xanthomatosis with chenodeoxycholic acid. N Engl J
Med 1984; 311(26): 1649-52.
[88] Yahalom G, Tsabari R, Molshatzki N, et al. Neurological outcome
in cerebrotendinous xanthomatosis treated with chenodeoxycholic
acid: Early versus late diagnosis. Clin Neuropharmacol 2013;
36(3): 78-83.
Current and Promising Therapies in Autosomal Recessive Ataxias CNS & Neurological Disorders - Drug Targets, 2018, Vol. 17, No. 3 171
[89] Huidekoper HH, Vaz FM, Verrips A, Bosch AM. Hepatotoxicity
due to chenodeoxycholic acid supplementation in an infant with
cerebrotendinous xanthomatosis: Implications for treatment. Eur J
Pediatr 2016; 175(1): 143-6.
[90] Alobaidy H. Recent advances in the diagnosis and treatment of
niemann-pick disease type C in children: A guide to early diagnosis
for the general pediatrician. Int J Pediatr 2015; 2015: 816593.
[91] Carstea ED, Morris JA, Coleman KG, et al. Niemann-Pick C1
disease gene: Homology to mediators of cholesterol homeostasis.
Science 1997; 277(5323): 228-31.
[92] Naureckiene S, Sleat DE, Lackland H, et al. Identificatio n of HE1
as the second gene of Niemann-Pick C disease. Science 2000;
290(5500): 2298-301.
[93] Wijburg FA, Sedel F, Pineda M, et al. Development of a suspicion
index to aid diagnosis of Niemann-Pick disease type C. Neurology
2012; 78(20): 1560-7.
[94] Lyseng-Williamson KA. Miglustat: A review of its use in Nie-
mann-Pick disease type C. Drugs 2014; 74(1): 61-74.
[95] Patterson MC, Vecchio D, Prady H, Abel L, Wraith JE. Miglustat
for treatment of Niemann-Pick C disease: A randomised controlled
study. Lancet Neurol 2007; 6(9): 765-72.
[96] Wraith JE, Vecchio D, Jacklin E, et al. Miglustat in adult and juve-
nile patients with Niemann-Pick disease type C: Lo ng-term data
from a clinical trial. Mol Genet Metab 2010; 99(4): 351-7.
[97] Pineda M, Wraith JE, Mengel E, et al. Miglustat in patients with
Niemann-Pick disease Type C (NP-C): A multicenter observational
retrospective cohort study. Mol Genet Metab 2009; 98(3): 243-9.
[98] Liu B, Turley SD, Burns DK, et al. Reversal of defective lysosomal
transport in NPC disease ame liorates liver dysfunction and neu-
rodegeneration in the npc1-/- mouse. Proc Natl Acad Sci USA
2009; 106(7): 2377-82.
[99] Liu B. Therapeutic potential of cyclodextrins in the treatment of
Niemann-Pick type C disease. Clin Lipidol 2012; 7(3): 289-301.
[100] Matsuo M, Togawa M, Hirabaru K, et al. Effects of cyclodextrin in
two patients with Niemann-Pick Type C disease. Mol Genet Metab
2013; 108(1): 76-81.
[101] Matsuo M, Shraishi K, Wada K, et al. Effects of intracerebroven-
tricular administration of 2-hydroxypropyl-beta-cyclodextrin in a
patient with Niemann-Pick Type C disease. Mol Genet Metab Rep
2014; 1: 391-400.
[102] Alam MS, Getz M, Haldar K. Chronic administration of an HDAC
inhibitor treats both neurological and systemic Niemann-Pick type
C disease in a mouse model. Sci Transl Med 2016; 8(326):
326ra23.
[103] Newton J, Hait NC, Maceyka M, et al. FTY720/fingolimod in-
creases NPC1 and NPC2 expression and reduces cholesterol and
sphingolipid accumulation in Niemann-Pick type C mutant fibro-
blasts. FASEB J 2017; 31(4): 1719-30.
[104] Pipalia NH, Cosner CC, Huang A, et al. Histone deacetylase inhibi-
tor treatment dramatically reduces cholesterol accumulation in
Niemann-Pick type C1 mutant human fibroblasts. Proc Natl Acad
Sci U S A 2011; 108(14): 5620-5.
[105] Munkacsi AB, Chen FW, Brinkman MA, et al. An "exacerbate-
reverse" stra tegy in yeast identifies histone deacetylase inhibition
as a correction for cholesterol and sphingolipid transport defects in
human Niemann-Pick type C disease. J Biol Chem 2011; 286(27):
23842-51.
[106] Xie C, Gong XM, Luo J, Li BL, Song BL. Intracardiac injection of
AAV9-NPC1 significantly ameliorates Purkinje cell death and be-
havioral abnormalities in mouse Niemann-Pick typ e C disease. J
Lipid Res. 2017; 58(3): 512-8.
[107] Bouchard JP, Barbeau A, Bouchard R, Bouchard RW. Autosomal
recessive spastic ataxia of Charlevoix-Saguenay. Can J Neurol Sci
1978; 5(1): 61-9.
[108] Bouhlal Y, Amouri R, El Euch-Fayeche G, Hentati F. Autosomal
recessive spastic ataxia of Charlevoix-Saguenay: An overview.
Parkinsonism Relat Disord 2011; 17(6): 418-22.
[109] Engert JC, Berube P, Mercier J, et al. ARSACS, a spastic ataxia
common in northeastern Quebec, is caused by mutations in a new
gene encoding an 11.5-kb ORF. Nat Genet 2000; 24(2): 120-5.
[110] Parfitt DA, Michael GJ, Vermeulen EG, et al. The ataxia protein
sacsin is a functional co-chaperone that protects against polyglu-
tamine-expanded ataxin-1. Hum Mol Genet 2009; 18(9): 1556-65.
[111] Picher-Martel V, Valdmanis PN, Gould PV, Julien JP, Dupré N.
From animal models to human disease: A genetic approach for per-
sonalized medicine in ALS. Acta Neuropathol Commun 2016 Jul
11; 4(1): 70.
[112] Singh NN, Howell MD, Androphy EJ, Singh RN. How the discov-
ery of ISS-N1 led to the first medical therapy for spinal muscular
atrophy. Gene Ther 2017; 24(9): 520-6.
DISCLAIMER: The above article has been published in Epub (ahead of print) on the basis of the materials provided by the author. The Edito-
rial Department reserves the right to make minor modifications for further improvement of the manuscript.
PMID: 29676235
... Oxidative stress has been suggested to be implicated in the pathogenesis of numerous neurodegenerative diseases, including hereditary ataxias [12]. Many studies have been conducted to validate the therapeutic roles of antioxidants in ARCAs [13][14][15][16][17]. However, their findings showed that antioxidants have only marginal therapeutic benefits for the management of ARCAs. ...
... These antioxidants were found to be ineffective or only partially effective on the symptoms of ARCAs, which could be due to lack of understanding of their modes of action targeting oxidative stress. In addition, the focus is often on the clinical outcomes and not the underlying molecular mechanisms [16]. To date, no systematic review has focused on the therapeutic roles of antioxidants and their associated modes of action for the management of ARCAs. ...
... AOA1 is the most common form of ARCA in the Japanese population. In Portugal, AOA is the second most common form of ARCA after FRDA [16] with an overall prevalence estimated to be 0.41 per 100,000 in which 3.6% of ARCA cases are AOA1 [36]. ...
Article
Full-text available
Autosomal recessive cerebellar ataxias (ARCAs) are a heterogeneous group of rare neu-rodegenerative inherited disorders. The resulting motor incoordination and progressive functional disabilities lead to reduced lifespan. There is currently no cure for ARCAs, likely attributed to the lack of understanding of the multifaceted roles of antioxidant defense and the underlying mechanisms. This systematic review aims to evaluate the extant literature on the current developments of therapeutic strategies that target oxidative stress for the management of ARCAs. We searched Pub-Med, Web of Science, and Science Direct Scopus for relevant peer-reviewed articles published from 1 January 2016 onwards. A total of 28 preclinical studies fulfilled the eligibility criteria for inclusion in this systematic review. We first evaluated the altered cellular processes, abnormal signaling cascades , and disrupted protein quality control underlying the pathogenesis of ARCA. We then examined the current potential therapeutic strategies for ARCAs, including aromatic, organic and pharmacological compounds, gene therapy, natural products, and nanotechnology, as well as their associated antioxidant pathways and modes of action. We then discussed their potential as antioxi-dant therapeutics for ARCAs, with the long-term view toward their possible translation to clinical practice. In conclusion, our current understanding is that these antioxidant therapies show promise in improving or halting the progression of ARCAs. Tailoring the therapies to specific disease stages could greatly facilitate the management of ARCAs.
... Nevertheless, the results indicated that these antioxidants only partially alleviated symptoms of ICAs. This limitation may be because of the emphasis on clinical outcomes rather than a comprehensive understanding of the underlying molecular mechanisms associated with their approach to addressing oxidative stress (Picher-Martel and Dupre, 2018;Lew et al., 2022). The cause of ICAs is diverse (Coarelli et al., 2023). ...
... K + channels are important in neuronal excitability, neurotransmitter release, neuroinflammation, and synaptic transmission in PD pathology. In dopaminergic neurons, voltage-dependent K + currents mediate repolarizing action potentials and fine-tune pacemaker firing rates (Braga Neto et al., 2016;Picher-Martel and Dupre, 2018). Voltagegated Na + channels modulate pacemaker frequency (Chen et al., 2012;Sarva and Shanker, 2014). ...
Article
Full-text available
Numerous neurodegenerative diseases result from altered ion channel function and mutations. The intracellular redox status can significantly alter the gating characteristics of ion channels. Abundant neurodegenerative diseases associated with oxidative stress have been documented, including Parkinson’s, Alzheimer’s, spinocerebellar ataxia, amyotrophic lateral sclerosis, and Huntington’s disease. Reactive oxygen and nitrogen species compounds trigger posttranslational alterations that target specific sites within the subunits responsible for channel assembly. These alterations include the adjustment of cysteine residues through redox reactions induced by reactive oxygen species (ROS), nitration, and S-nitrosylation assisted by nitric oxide of tyrosine residues through peroxynitrite. Several ion channels have been directly investigated for their functional responses to oxidizing agents and oxidative stress. This review primarily explores the relationship and potential links between oxidative stress and ion channels in neurodegenerative conditions, such as cerebellar ataxias and Parkinson’s disease. The potential correlation between oxidative stress and ion channels could hold promise for developing innovative therapies for common neurodegenerative diseases.
... (2) A pesar de ser esta una enfermedad neurodegenerativa fatal, en los últimos años han surgido un grupo de estrategias, con el objetivo de modificar el curso de la misma, como es el caso del miglustac que hace más lenta su progresión aunque tiene efectos secundarios que en ocasiones obliga a suspender su uso. (1,14,15,16,17,18,19) El objetivo de este trabajo es contribuir al conocimiento de esta rara enfermedad neurovisceral progresiva de curso fatal. ...
... El miglustad ha sido usado para prevenir la acumulación de glucosilceramida en pacientes con formas moderadas a severas de Gaucher tipo 1 y del Nieman Pick tipo C. En esta paciente no se observaron resultados alentadores con este fármaco, quizá debido a lo ya avanzado de la enfermedad. (14,15,16,17,18,19) Se concluye que el deterioro neurológico progresivo, la cataplejía gelástica, paresia de la mirada vertical y la esplenomegalia, unido a los resultados del medulograma y el estudio genético permitieron el diagnóstico de esta entidad. ...
Article
Full-text available
Introducción: La enfermedad de Niemann-Pick tipo C es una enfermedad poco frecuente, autosómica recesiva, caracterizada por el depósito de lípidos a nivel lisosomal, que, a pesar de ser tratable, es mortal en todos los casos y representa una importante carga para los pacientes y sus familias. Objetivo: Contribuir al conocimiento de esta rara enfermedad neurovisceral progresiva de curso fatal. Presentación del caso: Se trata de una niña de 7 años de edad, que a los 2 años asistió a consulta por trastornos de la marcha, con deterioro progresivo de esta, así como del lenguaje y el comienzo de crisis epilépticas. Evolutivamente presentó cataplejías gelásticas, paresia de la mirada vertical y esplenomegalia. Estos elementos clínicos evolutivos fueron lo suficientemente distintivos para orientar la sospecha clínica y las investigaciones necesarias para llegar al diagnóstico definitivo de la enfermedad. Con la confirmación de que se trataba de la enfermedad de Niemann-Pick tipo C, se comenzó tratamiento con miglustad a dosis de 100 mg dos veces al día. Conclusiones: El deterioro neurológico progresivo, la cataplejía gelástica, la paresia de la mirada vertical y la esplenomegalia, unido a los resultados del medulograma y el estudio genético permitieron el diagnóstico de esta entidad.
... (2) A pesar de ser esta una enfermedad neurodegenerativa fatal, en los últimos años han surgido un grupo de estrategias, con el objetivo de modificar el curso de la misma, como es el caso del miglustac que hace más lenta su progresión aunque tiene efectos secundarios que en ocasiones obliga a suspender su uso. (1,14,15,16,17,18,19) El objetivo de este trabajo es contribuir al conocimiento de esta rara enfermedad neurovisceral progresiva de curso fatal. ...
... El miglustad ha sido usado para prevenir la acumulación de glucosilceramida en pacientes con formas moderadas a severas de Gaucher tipo 1 y del Nieman Pick tipo C. En esta paciente no se observaron resultados alentadores con este fármaco, quizá debido a lo ya avanzado de la enfermedad. (14,15,16,17,18,19) Se concluye que el deterioro neurológico progresivo, la cataplejía gelástica, paresia de la mirada vertical y la esplenomegalia, unido a los resultados del medulograma y el estudio genético permitieron el diagnóstico de esta entidad. ...
Article
Full-text available
Resumen Introducción: La enfermedad de Niemann-Pick tipo C es una enfermedad poco frecuente, autosómica recesiva, caracterizada por el depósito de lípidos a nivel lisosomal, que, a pesar de ser tratable, es mortal en todos los casos y representa una importante carga para los pacientes y sus familias. Objetivo: Contribuir al conocimiento de esta rara enfermedad neurovisceral progresiva de curso fatal. Presentación del caso : Se trata de una niña de 7 años de edad, que a los 2 años asistió a consulta por trastornos de la marcha, con deterioro progresivo de esta, así como del lenguaje y el comienzo de crisis epilépticas. Evolutivamente presentó cataplejías gelásticas, paresia de la mirada vertical y esplenomegalia. Estos elementos clínicos evolutivos fueron lo suficientemente distintivos para orientar la sospecha clínica y las investigaciones necesarias para llegar al diagnóstico definitivo de la afección. Con la confirmación de que se trataba de una enfermedad de Niemann-Pick tipo C, se comenzó tratamiento con Miglustad a dosis de 100 mg dos veces al día. Conclusiones: El deterioro neurológico progresivo, la cataplejía gelástica, la paresia de la mirada vertical y la esplenomegalia, unido a los resultados del medulograma y el estudio genético permitieron el diagnóstico de esta entidad.
... Since ARSACS is a monogenic disorder characterized by SACS loss-of-function mutations, therapeutic options for loss-of-function diseases are significantly harder to develop when compared with their gain-of-function counterparts, with cancer therapeutics among the exceptions since tumor suppressor genes loss-of-function is exploited to develop synthetic lethality-based cancer drugs to target parallel genes that are activated upon tumor suppressor gene disruptions [47]. To date, pharmacological treatments for ARSACS management are scarce and only alleviate disease symptoms without addressing its underlying molecular mechanisms, with baclofen as an example used to manage spasticity and minimize tendon shortening [48]. ...
Article
Full-text available
Autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS) is a rare early-onset neurodegenerative disease caused by mutations in the SACS gene, encoding Sacsin. Initial functional annotation of Sacsin was based on sequence homology, with subsequent experiments revealing the Sacsin requirement for regulating mitochondrial dynamics, along with its domains involved in promoting neurofilament assembly or resolving their bundling accumulations. ARSACS phenotypes associated with SACS loss-of-function are discussed, and how advancements in ARSACS disease models and quantitative omics approaches can improve our understanding of ARSACS pathological attributes. Lastly in the perspectives section, we address gene correction strategies for monogenic disorders such as ARSACS, along with their common delivery methods, representing a hopeful area for ARSACS therapeutics development.
... This compound has been suggested as a therapeutic agent to improve brain functions in ARSACS disease by triggering anti-apoptotic and anti-inflammatory responses [4]. Baclofen is instead a synaptic reflex blocking agent that has been exploited as a therapeutic agent to reduce the symptoms in ARSACS patients, in terms of spasticity and muscle contractions [5]. Besides traditional pharmaceutical strategies, physical activity based on rehabilitation approaches has been also proposed to provide a symptomatic treatment in ARSACS patients [6]. ...
Article
Full-text available
Autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS) is a neurological disease characterized by autosomal recessive mutations in the sacsin gene (SACS), that cause in patients progressive cerebellar atrophy, damage of the peripheral nerves, and cognitive impairment. No effective therapies have been proposed for ARSACS, even if some evidences suggest that powerful antioxidant agents can be considered as a therapeutic tool. Resveratrol (Res) is a natural polyphenol compound derived from vegetal sources, the application of which in biomedicine is increasing in the latest years owing to its significant therapeutic effects, in particular in neurodegenerative diseases. In this study, we provide evidences about its potential exploitation in the treatment of ARSACS. Because of the low solubility of Res in physiological media, a nanoplatform based on nanostructured lipid carriers is proposed for its encapsulation and delivery. Resveratrol-loaded nanostructured lipid carriers (Res-NLCs) have been synthetized, characterized, and tested on healthy and ARSACS patient fibroblasts. Nanovectors displayed optimal stability and biocompatibility, and excellent antioxidant and anti-inflammatory activities. A comprehensive investigation at gene (with real-time quantitative RT-PCR) and protein (with proteomics) level demonstrated the therapeutic potential of Res-NLCs, encouraging future investigations on pre-clinical models.
Article
Full-text available
Background Autosomal recessive cerebellar ataxias (ARCA) are a group of rare inherited disorders characterized by degeneration or abnormal development of the cerebellum. Autosomal recessive spastic ataxia of Charlevoix–Saguenay (ARSACS) is one of the most prevalent in Europe. Objectives The aim of this study is to provide a better understanding of the manifestations and impacts of ARSACS. Methods A systematic review of the literature was conducted, followed by a qualitative study using semistructured interviews and discussion groups to obtain the experience of people affected. Results According to the PROMIS framework, the results show manifestations and impacts in three components of health: physical, mental, and social. Fatigue and struggles with balance and dexterity are the physical manifestations of the disease most often cited by participants. Negative affects such as frustration and depression are among the mental health impacts with some loss in cognitive abilities. Social health is the least documented component; nonetheless, people with the disease report significant impacts in terms of social relationships, activities and work. Conclusions These findings shed new light on the experience of people with recessive ataxia and identify key aspects to assess to improve their overall health.
Preprint
Full-text available
Background. Autosomal recessive cerebellar ataxias (ARCA) are a group of rare inherited disorders characterized by degeneration or abnormal development of cerebellum. Autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS) is the second most prevalent in Europe. Objectives. The aim of this study is to provide a better understanding of manifestations and impacts of ARSACS. Methods. A systematic review of literature has been conducted followed by a qualitative study using semi-structured interviews and discussion groups to get the experience of people affected. Results. According to the PROMIS framework, results show manifestations and impacts in three components of health: physical, mental, and social. Fatigue and struggles with balance and dexterity are the are the physical manifestations of the disease most often cited by participants. Negative affects like frustration and depression are among the mental health impacts with some loss in cognitive abilities. Social health is the least documented component; nonetheless, people with the disease report significant impacts in terms of social relationships, activities and work. Conclusions. These findings shed a new light on the experience of people with recessive ataxia and identify key aspects to assess to improve their overall health.
Article
The autosomal recessive cerebellar ataxias (ARCAs) compose a clinically and genetically heterogeneous group of neurodegenerative diseases characterized by prominent cerebellar ataxia, dysmetria, dysarthria, and nystagmus that are inherited in an autosomal recessive fashion. The diagnosis of ARCAs is challenging because of their low prevalence, poor medical recognition, and heterogeneous clinical presentation with many overlapping features between entities. There currently exist no disease-modifying therapies for most ARCAs, and treatment is mainly symptomatic, aimed at prolonging independence and maintaining quality of life. As knowledge of the common pathogenic pathways underlying several ARCAs grows, so do these pathways to target with new drugs. Chelation or enzyme replacement therapies are available for some specific ataxias caused by amenable metabolic alterations. A large number of drug trials are ongoing and aim to identify new therapeutic approaches to expand the options in our repertoire. Improved protocols of motor rehabilitation and noninvasive cerebellar stimulation were shown to delay disease progression and maintain quality of life. Furthermore, recent progress in gene and molecular targeting therapies are rapidly expanding and hold promise for repairing defective genes. Neurotransplantation of grafted stem cells, which is still at the experimental preclinical stage, has opened new therapeutic strategies aimed to delay cell degeneration and facilitate compensatory functions. This article is an overview of the current management and treatment strategies with an emphasis on promising perspectives for patients with ARCAs.
Article
Background: Sialuria is a rare inborn error of metabolism caused by excessive synthesis of sialic acid due to the mutation in the binding site of the cytidine monophosphate-sialic acid of UDPGlcNAc 2-Epimerase/ManNAc Kinase (GNE/MNK). Objective: This is the first study investigating the molecular basis of neuronal disorders exhibiting sialuria in Pakistani children/adolescents. Methods: The current study genotyped GNE SNPs rs121908621, rs121908622 and rs121908623 by using PCR, RFLP, and DNA sequencing methods. Socioeconomic and clinical histories were also recorded. Results: Our data suggest that clinical symptoms and financial status play a significant role in conferring sialuria related Intellectual Disability (ID). SNP: rs121908623 showed G/A substitution (R263Q) in the GNE gene. Conclusion: We have identified one case study in Pakistan, so this makes our research a leap forward towards the identification of the 10th case study worldwide.
Article
Full-text available
Spinal muscular atrophy (SMA), a prominent genetic disease of infant mortality, is caused by low levels of survival motor neuron (SMN) protein owing to deletions or mutations of the SMN1 gene. SMN2, a nearly identical copy of SMN1 present in humans, cannot compensate for the loss of SMN1 due to predominant skipping of exon 7 during pre-mRNA splicing. With the recent FDA approval of nusinersen (Spinraza™), the potential for correction of SMN2 exon 7 splicing as a SMA therapy has been affirmed. Nusinersen is an antisense oligonucleotide that targets intronic splicing silencer N1 (ISS-N1) discovered in 2004 at the University of Massachusetts Medical School. ISS-N1 has emerged as the model target for testing the therapeutic efficacy of antisense oligonucleotides using different chemistries as well as different mouse models of SMA. Here we provide a historical account of events that led to the discovery of ISS-N1 and describe the impact of independent validations that raised the profile of ISS-N1 as one of the most potent antisense targets for the treatment of a genetic disease. Recent approval of nusinersen provides a much-needed boost for antisense technology that is just beginning to realize its potential. Beyond treating SMA, the ISS-N1 target offers myriad potentials for perfecting various aspects of the nucleic-acid-based technology for the amelioration of the countless number of pathological conditions.Gene Therapy accepted article preview online, 09 May 2017. doi:10.1038/gt.2017.34.
Article
Full-text available
Niemann-Pick type C (NPC) disease is a fatal neurodegenerative disorder caused by mutations in NPC1 or NPC2 with decreased functions leading to lysosomal accumulation of cholesterol and sphingolipids. FTY720/fingolimod, used for treatment of multiple sclerosis, is phosphorylated by nuclear sphingosine kinase 2 and its active phosphorylated form (FTY720-P) is an inhibitor of class I histone deacetylases. In this study, administration of clinically relevant doses of FTY720 to mice increased expression of NPC1 and -2 in brain and liver and decreased cholesterol in an SphK2-dependent manner. FTY720 greatly increased expression of NPC1 and -2 in human NPC1 mutant fibroblasts that correlated with formation of FTY720-P and significantly reduced the accumulation of cholesterol and glycosphingolipids. In agreement with this finding, FTY720 pretreatment of human NPC1 mutant fibroblasts restored transport of the cholera toxin B subunit, which binds ganglioside GM1, to the Golgi apparatus. Together, these findings suggest that FTY720 administration can ameliorate cholesterol and sphingolipid storage and trafficking defects in NPC1 mutant fibroblasts. Because neurodegeneration is the main clinical feature of NPC disease, and FTY720 accumulates in the CNS and has several advantages over available histone deacetylase inhibitors now in clinical trials, our work provides a potential opportunity for treatment of this incurable disease.-Newton, J., Hait, N. C., Maceyka, M., Colaco, A., Maczis, M., Wassif, C. A., Cougnoux, A., Porter, F. D., Milstien, S., Platt, N., Platt, F. M., Spiegel, S. FTY720/fingolimod increases NPC1 and NPC2 expression and reduces cholesterol and sphingolipid accumulation in Niemann-Pick type C mutant fibroblasts.
Article
Full-text available
Niemann-Pick type C (NPC) disease is a fatal, inherited neurodegenerative disorder caused by loss-of-function mutations in the NPC1 or NPC2 gene. There is no effective way to treat NPC disease. In this study, we used adeno-associated virus (AAV) serotype 9 to deliver a functional NPC1 gene systemically into NPC1-/- mice at postnatal day 4 (P4). One single AAV9-NPC1 injection resulted in robust NPC1 expression in various tissues including the brain, heart and lung. Strikingly, AAV9-mediated NPC1 delivery significantly promoted Purkinje cell survival, restored locomotor activity and coordination, as well as increased the lifespan of NPC1-/- mice. Our work suggests that AAV-based gene therapy is a promising means to treat NPC disease.
Article
Full-text available
Definition of the disease: Ataxia telangiectasia (A-T) is an autosomal recessive disorder primarily characterized by cerebellar degeneration, telangiectasia, immunodeficiency, cancer susceptibility and radiation sensitivity. A-T is often referred to as a genome instability or DNA damage response syndrome. Epidemiology: The world-wide prevalence of A-T is estimated to be between 1 in 40,000 and 1 in 100,000 live births. Clinical description: A-T is a complex disorder with substantial variability in the severity of features between affected individuals, and at different ages. Neurological symptoms most often first appear in early childhood when children begin to sit or walk. They have immunological abnormalities including immunoglobulin and antibody deficiencies and lymphopenia. People with A-T have an increased predisposition for cancers, particularly of lymphoid origin. Pulmonary disease and problems with feeding, swallowing and nutrition are common, and there also may be dermatological and endocrine manifestations. Etiology: A-T is caused by mutations in the ATM (Ataxia Telangiectasia, Mutated) gene which encodes a protein of the same name. The primary role of the ATM protein is coordination of cellular signaling pathways in response to DNA double strand breaks, oxidative stress and other genotoxic stress. Diagnosis: The diagnosis of A-T is usually suspected by the combination of neurologic clinical features (ataxia, abnormal control of eye movement, and postural instability) with one or more of the following which may vary in their appearance: telangiectasia, frequent sinopulmonary infections and specific laboratory abnormalities (e.g. IgA deficiency, lymphopenia especially affecting T lymphocytes and increased alpha-fetoprotein levels). Because certain neurological features may arise later, a diagnosis of A-T should be carefully considered for any ataxic child with an otherwise elusive diagnosis. A diagnosis of A-T can be confirmed by the finding of an absence or deficiency of the ATM protein or its kinase activity in cultured cell lines, and/or identification of the pathological mutations in the ATM gene. Differential diagnosis: There are several other neurologic and rare disorders that physicians must consider when diagnosing A-T and that can be confused with A-T. Differentiation of these various disorders is often possible with clinical features and selected laboratory tests, including gene sequencing. Antenatal diagnosis: Antenatal diagnosis can be performed if the pathological ATM mutations in that family have been identified in an affected child. In the absence of identifying mutations, antenatal diagnosis can be made by haplotype analysis if an unambiguous diagnosis of the affected child has been made through clinical and laboratory findings and/or ATM protein analysis. Genetic counseling: Genetic counseling can help family members of a patient with A-T understand when genetic testing for A-T is feasible, and how the test results should be interpreted. Management and prognosis: Treatment of the neurologic problems associated with A-T is symptomatic and supportive, as there are no treatments known to slow or stop the neurodegeneration. However, other manifestations of A-T, e.g. immunodeficiency, pulmonary disease, failure to thrive and diabetes can be treated effectively.
Article
Full-text available
Background: The autosomal recessive ataxias are a heterogeneous group of disorders that are characterized by complex neurological features in addition to progressive ataxia. Hyperkinetic movement disorders occur in a significant proportion of patients, and may sometimes be the presenting motor symptom. Presentations with involuntary movements rather than ataxia are diagnostically challenging, and are likely under-recognized. Methods: A PubMed literature search was performed in October 2015 utilizing pairwise combinations of disease-related terms (autosomal recessive ataxia, ataxia-telangiectasia, ataxia with oculomotor apraxia type 1 (AOA1), ataxia with oculomotor apraxia type 2 (AOA2), Friedreich ataxia, ataxia with vitamin E deficiency), and symptom-related terms (movement disorder, dystonia, chorea, choreoathetosis, myoclonus). Results: Involuntary movements occur in the majority of patients with ataxia-telangiectasia and AOA1, and less frequently in patients with AOA2, Friedreich ataxia, and ataxia with vitamin E deficiency. Clinical presentations with an isolated hyperkinetic movement disorder in the absence of ataxia include dystonia or dystonia with myoclonus with predominant upper limb and cervical involvement (ataxia-telangiectasia, ataxia with vitamin E deficiency), and generalized chorea (ataxia with oculomotor apraxia type 1, ataxia-telangiectasia). Discussion: An awareness of atypical presentations facilitates early and accurate diagnosis in these challenging cases. Recognition of involuntary movements is important not only for diagnosis, but also because of the potential for effective targeted symptomatic treatment.
Article
Full-text available
Friedreich ataxia (FRDA) is a progressive neurodegenerative disease caused by deficiency of frataxin protein, with the primary sites of pathology being the large sensory neurons of the dorsal root ganglia and the cerebellum. FRDA is also often accompanied by severe cardiomyopathy and diabetes mellitus. Frataxin is important in mitochondrial iron-sulfur cluster (ISC) biogenesis and low-frataxin expression is due to a GAA repeat expansion in intron 1 of the FXN gene. FRDA cells are genomically unstable, with increased levels of reactive oxygen species and sensitivity to oxidative stress. Here we report the identification of elevated levels of DNA double strand breaks (DSBs) in FRDA patient and YG8sR FRDA mouse model fibroblasts compared to normal fibroblasts. Using lentivirus FXN gene delivery to FRDA patient and YG8sR cells, we obtained long-term overexpression of FXN mRNA and frataxin protein levels with reduced DSB levels towards normal. Furthermore, γ-irradiation of FRDA patient and YG8sR cells revealed impaired DSB repair that was recovered on FXN gene transfer. This suggests that frataxin may be involved in DSB repair, either directly by an unknown mechanism, or indirectly via ISC biogenesis for DNA repair enzymes, which may be essential for the prevention of neurodegeneration.Gene Therapy advance online publication, 20 October 2016; doi:10.1038/gt.2016.61.
Article
Full-text available
Amyotrophic Lateral Sclerosis (ALS) is the most frequent motor neuron disease in adults. Classical ALS is characterized by the death of upper and lower motor neurons leading to progressive paralysis. Approximately 10 % of ALS patients have familial form of the disease. Numerous different gene mutations have been found in familial cases of ALS, such as mutations in superoxide dismutase 1 (SOD1), TAR DNA-binding protein 43 (TDP-43), fused in sarcoma (FUS), C9ORF72, ubiquilin-2 (UBQLN2), optineurin (OPTN) and others. Multiple animal models were generated to mimic the disease and to test future treatments. However, no animal model fully replicates the spectrum of phenotypes in the human disease and it is difficult to assess how a therapeutic effect in disease models can predict efficacy in humans. Importantly, the genetic and phenotypic heterogeneity of ALS leads to a variety of responses to similar treatment regimens. From this has emerged the concept of personalized medicine (PM), which is a medical scheme that combines study of genetic, environmental and clinical diagnostic testing, including biomarkers, to individualized patient care. In this perspective, we used subgroups of specific ALS-linked gene mutations to go through existing animal models and to provide a comprehensive profile of the differences and similarities between animal models of disease and human disease. Finally, we reviewed application of biomarkers and gene therapies relevant in personalized medicine approach. For instance, this includes viral delivering of antisense oligonucleotide and small interfering RNA in SOD1, TDP-43 and C9orf72 mice models. Promising gene therapies raised possibilities for treating differently the major mutations in familial ALS cases.
Article
Full-text available
Infantile Refsum disease (IRD) is a rare autosomal recessive disorder of peroxisome biogenesis characterized by generalized peroxisomal metabolic dysfunction, including accumulation of very long-chain fatty acids (VLCFAs) and phytanic acid (PA), as well as decreased plasmalogen contents (PL). An effective therapy for this intractable disease has not been established, and only supportive management with docosahexaenoic acid supplementation and low PA diet has been reported so far. A boy of 3 years and 8 months presented with facial dysmorphism, transaminitis, and psychomotor retardation. Biochemical analysis showed elevated PA and VLCFAs, with reduced PL in the serum. Immunofluorescence study of fibroblasts from the patient indicated a mosaic pattern of catalase-positive and -negative particles, and molecular analysis revealed compound heterozygous mutations of PEX6 The failure of medical management to prevent the progression of clinical symptoms and abnormal biochemistry prompted us to consider liver transplantation (LT). With the chances of receiving a deceased donor liver being poor, we performed a living-donor LT from the patient's heterozygous mother. At 6-month follow-up, the patient's serum PA levels had normalized. VLCFAs and PL levels had declined and increased, respectively. To the best of our knowledge, this is the second reported case in which IRD was treated by living-donor LT by using a heterozygous donor. Only long-term follow-up will reveal if there is any clinical improvement in the present case. With the liver being a major site for peroxisomal pathways, its replacement by LT may work as a form of partial enzyme therapy for patients with IRD.
Article
Ataxia Telangiectasia (A-T) is a rare incurable neurodegenerative disease caused by biallelic mutations in the ATM gene. The lack of a functional ATM kinase leads to a pleiotropic phenotype, and oxidative stress is thought to have a crucial role in the complex physiopathology. Recently, steroids have been shown to reduce the neurological symptoms of the disease, but the molecular mechanism of this effect is largely unknown. We demonstrated that dexamethasone treatment of A-T lymphoblastoid cells increases the content of two of the most abundant antioxidants, GSH and NADPH, by up to 30%. Dexamethasone promoted the nuclear accumulation of the transcription factor NRF2 to drive expression of antioxidant pathways involved in glutathione synthesis and NADPH production. The latter effect was via G6PD activation, as confirmed by increased enzyme activity and enhancement of the pentose phosphate pathway rate. This evidence indicates that glucocorticoids are able to potentiate antioxidant defenses to counteract oxidative stress in Ataxia Telangiectasia, and reveal an unexpected role for dexamethasone in redox homeostasis and cellular antioxidant activity. This article is protected by copyright. All rights reserved.
Article
Histone deacetylase inhibitors (HDACi) are approved for treating rare cancers and are of interest as potential therapies for neurodegenerative disorders. We evaluated a triple combination formulation (TCF) comprising the pan-HDACi vorinostat, the caging agent 2-hydroxypropyl-β-cyclodextrin (HPBCD), and polyethylene glycol (PEG) for treating a mouse model (the Npc1nmf164 mouse) of Niemann-Pick type C (NPC) disease, a difficult-to-treat cerebellar disorder. Vorinostat alone showed activity in cultured primary cells derived from Npc1nmf164 mice but did not improve animal survival.However, low-dose, once-weekly intraperitoneal injections of the TCF containing vorinostat increased histone acetylation in the mouse brain, preserved neurites and Purkinje cells, delayed symptoms of neurodegeneration, and extended mouse life span from 4 to almost 9 months. We demonstrate that the TCF boosted the ability of HDACi to cross the blood-brain barrier and was not toxic even when used long term. Further, the TCF enabled dose reduction, which has been amajor challenge in HDACi therapy. TCF simultaneously treats neurodegenerative and systemic symptoms of Niemann-Pick type C disease in a mouse model. Copyright 2016 by the American Association for the Advancement of Science; all rights reserved.