Skip to main content
Erschienen in: Journal of Neurology 8/2015

Open Access 01.08.2015 | Original Communication

Frequency of rare recessive mutations in unexplained late onset cerebellar ataxia

verfasst von: M. J. Keogh, H. Steele, K. Douroudis, A. Pyle, J. Duff, R. Hussain, T. Smertenko, H. Griffin, M. Santibanez-Koref, R. Horvath, P. F. Chinnery

Erschienen in: Journal of Neurology | Ausgabe 8/2015

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Sporadic late onset cerebellar ataxia is a well-described clinical presentation with a broad differential diagnosis that adult neurologists should be familiar with. However, despite extensive clinical investigations, an acquired cause is identified in only a minority of cases. Thereafter, an underlying genetic basis is often considered, even in those without a family history. Here we apply whole exome sequencing to a cohort of 12 patients with late onset cerebellar ataxia. We show that 33 % of ‘idiopathic’ cases harbor compound heterozygous mutations in known ataxia genes, including genes not included on multi-gene panels, or primarily associated with an ataxic presentation.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s00415-015-7772-x) contains supplementary material, which is available to authorized users.
M. J. Keogh and H. Steele contributed equally to this work and R. Horvath and P. F. Chinnery contributed equally to this work.

Introduction

Adult onset cerebellar ataxia poses a considerable diagnostic challenge. Initial investigations focus on detecting degenerative, toxic, structural and inflammatory etiologies which together underlie around a third of cases [1]. Thereafter, molecular investigations for a monogenic basis of disease are often undertaken despite 80 % of patients having no relevant family history [2].
Current molecular investigations for sporadic cases echo that of familial forms, beginning with testing for trinucleotide repeat disorders, such as the spinocerebellar ataxias (SCA1, 2, 3, 6, 7 and 17), dentatorubral pallidoluysian atrophy (DRPLA) and Friedreich’s ataxia (FDR) in most centres [1]. However, this approach fails to identify a molecular diagnosis in 87–98 % of late onset sporadic cases [1, 3], and subsequent investigations are undertaken on a gene-by-gene basis, often at considerable time and expense.
The difficulty in establishing monogenic forms of disease using this approach is increasingly challenging given that at least 60 causative ataxia genes are reported [4]. Recent studies have therefore utilized next generation sequencing focusing on infantile or juvenile onset cases [5], or adult onset ataxia with a demonstrable family history [4]. Only two studies have described sub-sets of patients with sporadic onset adult disease, despite it being a major form of ataxia, and suggested that a molecular diagnosis can be reached in ~10 % of cases [4, 6]. Given this, we applied whole exome sequencing to a cohort of individuals with sporadic late onset ataxia.

Methods

Unrelated individuals with sporadic ataxia beginning at 30 years of age or over were identified from routine referrals to our regional neurogenetic service, in Newcastle upon Tyne, England.
Acquired causes of ataxia were excluded and all participants had negative genetic testing for SCA 1, 2, 3, 6, 7, 17, DRPLA and Friedreich’s Ataxia (FA). In addition, all adult males had negative FMR1 testing.
Blood genomic DNA was fragmented, exome enriched and sequenced (Nextera Rapid Exome Capture 37 Mb and HiSeq 2000, 100 bp paired-end reads). In-house bioinformatic analysis included alignment to UCSC hg19, using BWA as aligner and GATK to detect SNV and INDELS across all samples using standard filtering parameters according to GATK Best Practise Recommendations [7] (see supplementary methods). Further analysis was performed on variants with a minor allele frequency <0.005 in several reference databases and 302 unrelated in-house controls (see supplementary methods). Rare heterozygous, homozygous and compound heterozygous variants were defined, and protein altering and/or putative ‘disease causing’ mutations as predicted by at least three out of four software programmes were included. Pathogenicity was defined in accordance with American College of Medical Genetic guidelines (see supplementary methods). Genes known or suggested to cause ataxia as a primary or secondary phenotype in humans from two suggested clinical panels [4, 8] together with additional genes in which ataxia may result as part of the phenotype (list-supplementary methods) were assessed for variants according to the above criteria, and confirmed by Sanger sequencing (supplementary methods).
Variants were defined using a priori criteria: (1) confirmed pathogenic: dominant disordersvariant previously shown to cause ataxia in humans; recessive disorderseither 2 variants previously shown to cause ataxia in humans; or 1 pathogenic variant with a second variant predicted to affect protein function by at least 3 of 4 prediction algorithms (SIFT, Polyphen2, Mutation Taster, LRT), or through frameshift or truncation. (2) Probable pathogenic: dominant and recessive disorders—variants in known genes causing ataxia in humans and predicted to affect protein function by at least three of four prediction algorithms; (3) uncertain significance: dominant and recessive disorders—variants predicted to affect protein function with weak evidence that gene alteration causes ataxia in humans.
The study was granted ethical approval from a Research Ethics Committee based in the North of England.

Results

Population

Twelve Caucasian individuals of British origin (5 male) with no known consanguinity were included (Table 1). Mean age at disease onset was 46.7 years (SD 11; range 30–70 years). Mean disease duration was 16.6 years (SD 6.9; range 6–30 years). For one patient, the disease duration fell within the range expected for multi-system atrophy [9]. This patient had a normal DaTscan and autonomic function tests. Three individuals had CSF examination with negative oligoclonal bands. Five had nerve conduction studies; two of which were abnormal. Detailed clinical features and the results of clinical investigations are shown in Table 1.
Table 1
Clinical features of the 12 patients in the cohort
Patient no., sex
Age (years)
Age onset (years)
Disease duration (years)
Presenting symptom
Gait ataxia
Limb ataxia
Ocular signs
Additional neurological features
Other features
MRI
LP
NCS/EMG
Other investigations
Muscle biopsy
Other negative molecular investigations
1, F
63
40
23
Slowly progressive midline and appendicular ataxic syndrome
+++
++
Early CPEO
Dysmetric pursuit
Broken saccades
Dysphagia, spastic bladder
Lower limb spasticity
None
CA
Normal
−OCB
Bilateral CTS (CTS study only)
 
Normal IHC
No mtDNA deletions
FMR1
2, F
47
30
17
Slowly progressive spastic ataxic syndrome
++ (Fr)
+
CPEO
Temporal optic disc pallor
Jerky pursuit
Hypometric saccades
Spastic lower limbs
Brisk reflexes
None
Mild CA
Normal
−OCB
ND
 
Mild fibre size variation
Low Q10
Nil
3, F
57
45
12
Ataxia developed aged 45
+++
++
Slow saccades
Epilepsy aged 7
None
CA and parieto-occipital atrophy
ND
Normal
 
ND
FMR1
4, F
63
40
23
Slowly progressive midline cerebellar ataxia
++
+
GEN
TLE with ongoing infrequent focal seizures, no treatment
Cataracts (age 62)
CA
ND
ND
 
ND
POLG
MT-ATP6 & 8
5, F
55
35
20
Slowly progressive spastic ataxic syndrome
+++ (WhC)
++
Jerky pursuit
GEN
Hypometric saccades
Neurogenic bladder
Spastic ataxic gait
Brisk reflexes
Positive Babinski
None
CA
ND
ND
 
ND
SPG7
6, F
76
70
6
Progressive midline and appendicular ataxia
+++
++
GEN
Up and down beat nystagmus
Orthostatic tremor
Brisk reflexes
None
Mild CA
ND
ND
−DaT
Normal IHC
No mtDNA deletions
MT-ATP 6 & 8
7, M
71
60
11
Slowly progressive midline ataxia
+
Jerky ocular pursuit
GEN
None
None
CA
ND
Normal
 
ND
SPG7
MT-ATP 6 & 8
8, M
58
50
8
Midline ataxia
++
+
RAPD
OA
Jerky pursuit
GEN
Congenital hearing loss
Early dysphagia
Areflexia
None
CA
ND
SAN
 
Normal IHC
No mtDNA deletions
Normal Q10
POLG
WFS1
OPA1
MT-ATP 6 & 8
9, M
70
40
30
Pure midline ataxia
+ (stick)
None
None
None
CA
ND
ND
 
Normal IHC
Normal RCE
SCA12
mt.DNA LR-PCR
10, M
59
44
15
Pure midline ataxia
+++
+
None
Prominent dysarthria, choking
Brisk reflexes
None
CA
ND
ND
 
Normal Q10
SPG7
SCA8
SCA12
11, F
65
47
12
Pure midline ataxia
+++ (WhC)
+
Oscillopsia
Jerky pursuits
Horizontal nystagmus
Hypometric saccades
Dorsal root ganglionopathy
Neurogenic bladder
Distal wasting and weakness
Areflexia
Cataract, diabetes and short stature
Mild CA; high signal C3, 4 posterior columns; thin cord
−OCB
DRG
 
ND
POLG
SPG7
POLG2
PEO1
ANT1
mt.DNA LR-PCR
12, M
83
60
23
Midline ataxia
Early alcohol sensitivity
++ (stick)
+
Jerky pursuit
Coarse phasic nystagmus
Normal saccades
None
None
Mild CA
ND
ND
 
Patient declined
Nil
Presence or absence of symptoms are indicated by + or − symbol, respectively
AFTs autonomic function tests, CA Cerebellar atrophy, CPEO chronic progressive external ophthalmoplegia, CTS carpal tunnel syndrome, CVD cerebrovascular disease, DRG dorsal root ganglionopathy, Fr Frame, GEN gaze evoked nystagmus, IHC immunohistochemistry, ND not done, OA optic atrophy, OCB oligoclonal bands, PV periventricular, RAPD relative afferent pupillary defect, RCE respiratory chain enzyme, SVD small vessel disease, TLE temporal lobe epilepsy, WhC wheelchair, WM white matter

Diagnosis

We identified previously described pathogenic mutations in four of the 12 (33 %) patients in our cohort. All were present on confirmatory Sanger sequencing. No probable pathogenic variants were identified and variants of uncertain significance were found in an additional two cases (17 %). Findings are summarised in Table 2.
Table 2
Genetic variants of interest identified in the 12 patients
Pathogenic variants
Pt
Gene
Model
Exome seq identified variant (1)
rs#
MAF variant (1)
Exome seq identified variant (2)
rs#
MAF variant (2)
Variant pathogenicity prediction
ESP6500
1000 g
ESP6500
1000 g
1
SPG7
AR
c.1529C>T
p. Ala510Val
rs61755320
0.003463
0.0014
c. 1053dupC
p. Gly352fs
NA
0
0
(1) D:D:D:D
(2) NA
2
SPG7
AR
c.1529C>T
p. Ala510Val
rs61755320
0.003463
0.0014
c.233T>A
p. Leu78*
rs121918358
0.000077
0
(1) D:D:D:D
(2) Pathogenic
3
ANO10
AR
c.1843G>A
p. Asp615Asn
rs138000380
0.000231
0.0005
c. 132_133insT
p. Asp45fs
NA
0
0
(1) D:D:D:P
(2) NA
4
SYNE1
AR
c.9148C>G
p. Leu3050Val
rs117360770
0.002307
0.0018
c.1762delC
p. Leu588fs
NA
0.003435
0
(1) D:D:D:D
(2) NA
Variants of uncertain significance
5
SLC33A1
AD
c.433G>A
p. Gly145Ser
rs138283229
0.002461
0.0009
NA
 
NA
NA
D:D:D:D
6
PLEKHG4
AD
c.2251G>A
p. Asp751Asn
NA
0.000077
0
NA
 
NA
NA
D:D:N:D
Confirmed pathogenic: dominant disorders—variant previously shown to cause ataxia in humans; recessive disorders—either 2 variants previously shown to cause ataxia in humans; or 1 pathogenic variant with a second variant predicted to affect protein function by at least 3 of 4 prediction algorithms (SIFT, Polyphen2, Mutation Taster, LRT), through frameshift or truncation. Variants of uncertain significance: dominant and recessive disorders—variants predicted to affect protein function with weak evidence that gene alteration causes ataxia in humans
D pathogenic or deleterious, P polymorphism, NA not applicable N neutral (frameshift mutations considered pathogenic)

Discussion

We identified confirmed or probable pathogenic variants causing sporadic late onset ataxia in four patients (33 %) in our cohort. These findings are comparable to childhood/adolescent ataxia using targeted sequencing panels (40 %) [4] and whole exome sequencing (27 %) [5]. They are also significantly higher than previous data for adult onset cases using either panels or whole exome (both ~10 %) [4, 6].
We detected pathogenic variants in SPG7, SYNE1 and ANO10 (previously published by Balreira et al. [10]). Fogel et al. [6] also identified pathogenic variants in these genes (SPG7 (n = 2), SYNE1 (n = 3) and ANO10 (n = 1). The clinical features of these patients appear relatively homogenous between their and our study, with pure cerebellar ataxia beginning above the age of 40 for ANO10 and SYNE1 cases, and a more heterogeneous age of onset (<20–50) with additional neurological features including spasticity and a polyneuropathy in SPG7 cases [6]. Therefore, pathogenic mutations in these genes appear to be an important and frequently identified cause of late onset sporadic ataxia.
We used whole exome sequencing (WES) rather than targeted next generation ‘panels’, and it remains a contentious issue as to which is more appropriate in the investigation of neurogenetic disorders. WES enables greater genome coverage, and hence detection of pathogenic mutations in genes not considered as having ataxia as a primary phenotype. Our results highlight this as SPG7 was not covered by one ataxia panel [4], SYNE1 by another [8], and ANO10 was not included in either panel. WES however, may result in detection of unexpected findings such as pathogenic mutations predisposing to cancer or neurodegenerative disease, which must be considered and included in appropriate consent procedures. It must also be noted that neither WES nor targeted panels are appropriate to screen for genomic rearrangements or trinucleotide repeat sequences.
Determining pathogenicity can be challenging for heterozygous variants without a family history of disease and additional living family relatives for segregation analysis. In our cohort, we found heterozygous variants in SLC33A1 and PLEKHG4 in single cases (Table 2). Heterozygous mutations in SLC33A1 have been associated with spastic paraplegia (SPG42) with ataxia in a single family, and likewise, missense mutations in PLEKHG4 have been implicated in dominant late onset forms of spinocerebellar ataxia in Japanese individuals. Despite the rarity and putative pathogenicity of the variants in our patients, the lack of data to test segregation makes attributing pathogenicity difficult. As NGS begins to develop larger variant datasets in rare diseases it is vital to share such data through collaborative projects which may aid pathogenicity confirmation through the identification of the same or related variants in unrelated families with a similar phenotype.
In conclusion, we have demonstrated that application of WES to a cohort of unrelated individuals following exclusion of common trinucleotide repeat disorders establishes a molecular cause of disease in a third of cases. These findings have significant implications for clinical practise.

Acknowledgments

MJK is a Wellcome Trust Clinical Research Training Fellow. PFC is an Honorary Consultant Neurologist at Newcastle upon Tyne Foundation Hospitals NHS Trust, is a Wellcome Trust Senior Fellow in Clinical Science (101876/Z/13/Z), and a UK NIHR Senior Investigator. PFC receives additional support from the Wellcome Trust Centre for Mitochondrial Research (096919Z/11/Z), the Medical Research Council (UK) Centre for Translational Muscle Disease research (G0601943), and EU FP7 TIRCON, and the National Institute for Health Research (NIHR) Newcastle Biomedical Research Centre based at Newcastle upon Tyne Hospitals NHS Foundation Trust and Newcastle University. The views expressed are those of the author(s) and not necessarily those of the NHS, the NIHR or the Department of Health. RH was supported by the Medical Research Council (UK) (G1000848) and the European Research Council (309548).

Conflicts of interest

The authors report no conflicts of interest.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

Neuer Inhalt

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

Weitere Produktempfehlungen anzeigen
Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Abele M, Burk K, Schols L, Schwartz S, Besenthal I, Dichgans J, Zuhlke C, Riess O, Klockgether T (2002) The aetiology of sporadic adult-onset ataxia. Brain J Neurol 125:961–968CrossRef Abele M, Burk K, Schols L, Schwartz S, Besenthal I, Dichgans J, Zuhlke C, Riess O, Klockgether T (2002) The aetiology of sporadic adult-onset ataxia. Brain J Neurol 125:961–968CrossRef
2.
Zurück zum Zitat Muzaimi MB, Thomas J, Palmer-Smith S, Rosser L, Harper PS, Wiles CM, Ravine D, Robertson NP (2004) Population based study of late onset cerebellar ataxia in south east Wales. J Neurol Neurosurg Psychiatry 75:1129–1134PubMedCentralCrossRefPubMed Muzaimi MB, Thomas J, Palmer-Smith S, Rosser L, Harper PS, Wiles CM, Ravine D, Robertson NP (2004) Population based study of late onset cerebellar ataxia in south east Wales. J Neurol Neurosurg Psychiatry 75:1129–1134PubMedCentralCrossRefPubMed
3.
Zurück zum Zitat Pujana MA, Corral J, Gratacos M, Combarros O, Berciano J, Genis D, Banchs I, Estivill X, Volpini V (1999) Spinocerebellar ataxias in Spanish patients: genetic analysis of familial and sporadic cases. The Ataxia Study Group. Hum Genet 104:516–522CrossRefPubMed Pujana MA, Corral J, Gratacos M, Combarros O, Berciano J, Genis D, Banchs I, Estivill X, Volpini V (1999) Spinocerebellar ataxias in Spanish patients: genetic analysis of familial and sporadic cases. The Ataxia Study Group. Hum Genet 104:516–522CrossRefPubMed
4.
Zurück zum Zitat Nemeth AH, Kwasniewska AC, Lise S, Parolin Schnekenberg R, Becker EB, Bera KD, Shanks ME, Gregory L, Buck D, Zameel Cader M, Talbot K, de Silva R, Fletcher N, Hastings R, Jayawant S, Morrison PJ, Worth P, Taylor M, Tolmie J, O’Regan M, UK Ataxia Consortium, Valentine R, Packham E, Evans J, Seller A, Ragoussis J (2013) Next generation sequencing for molecular diagnosis of neurological disorders using ataxias as a model. Brain J Neurol 136:3106–3118CrossRef Nemeth AH, Kwasniewska AC, Lise S, Parolin Schnekenberg R, Becker EB, Bera KD, Shanks ME, Gregory L, Buck D, Zameel Cader M, Talbot K, de Silva R, Fletcher N, Hastings R, Jayawant S, Morrison PJ, Worth P, Taylor M, Tolmie J, O’Regan M, UK Ataxia Consortium, Valentine R, Packham E, Evans J, Seller A, Ragoussis J (2013) Next generation sequencing for molecular diagnosis of neurological disorders using ataxias as a model. Brain J Neurol 136:3106–3118CrossRef
5.
Zurück zum Zitat Sawyer SL, Schwartzentruber J, Beaulieu CL, Dyment D, Smith A, Warman Chardon J, Yoon G, Rouleau GA, Suchowersky O, Siu V, Murphy L, Hegele RA, Marshall CR, FORGE Canada Consortium, Bulman DE, Majewski J, Tarnopolsky M, Boycott KM (2014) Exome sequencing as a diagnostic tool for pediatric-onset ataxia. Hum Mutat 35:45–49PubMedCentralCrossRefPubMed Sawyer SL, Schwartzentruber J, Beaulieu CL, Dyment D, Smith A, Warman Chardon J, Yoon G, Rouleau GA, Suchowersky O, Siu V, Murphy L, Hegele RA, Marshall CR, FORGE Canada Consortium, Bulman DE, Majewski J, Tarnopolsky M, Boycott KM (2014) Exome sequencing as a diagnostic tool for pediatric-onset ataxia. Hum Mutat 35:45–49PubMedCentralCrossRefPubMed
6.
Zurück zum Zitat Fogel BL, Lee H, Deignan JL, Strom SP, Kantarci S, Wang X, Quintero-Rivera F, Vilain E, Grody WW, Perlman S, Geschwind DH, Nelson SF (2014) Exome sequencing in the clinical diagnosis of sporadic or familial cerebellar ataxia. JAMA Neurol 71:1237–1246CrossRefPubMed Fogel BL, Lee H, Deignan JL, Strom SP, Kantarci S, Wang X, Quintero-Rivera F, Vilain E, Grody WW, Perlman S, Geschwind DH, Nelson SF (2014) Exome sequencing in the clinical diagnosis of sporadic or familial cerebellar ataxia. JAMA Neurol 71:1237–1246CrossRefPubMed
7.
Zurück zum Zitat Van der Auwera GA, Carneiro MO, Hartl C, Poplin R, del Angel G, Levy-Moonshine A, Jordan T, Shakir K, Roazen D, Thibault J, Banks E, Garimella K, Altshuler D, Gabriel S, DePristo M (2013) From FastQ data to high-confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr Protoc Bioinformatics 43:11.10.11–11.10.33 Van der Auwera GA, Carneiro MO, Hartl C, Poplin R, del Angel G, Levy-Moonshine A, Jordan T, Shakir K, Roazen D, Thibault J, Banks E, Garimella K, Altshuler D, Gabriel S, DePristo M (2013) From FastQ data to high-confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr Protoc Bioinformatics 43:11.10.11–11.10.33
9.
Zurück zum Zitat Wenning GK, Geser F, Krismer F, Seppi K, Duerr S, Boesch S, Kollensperger M, Goebel G, Pfeiffer KP, Barone P, Pellecchia MT, Quinn NP, Koukouni V, Fowler CJ, Schrag A, Mathias CJ, Giladi N, Gurevich T, Dupont E, Ostergaard K, Nilsson CF, Widner H, Oertel W, Eggert KM, Albanese A, del Sorbo F, Tolosa E, Cardozo A, Deuschl G, Hellriegel H, Klockgether T, Dodel R, Sampaio C, Coelho M, Djaldetti R, Melamed E, Gasser T, Kamm C, Meco G, Colosimo C, Rascol O, Meissner WG, Tison F, Poewe W, European Multiple System Atrophy Study Group (2013) The natural history of multiple system atrophy: a prospective European cohort study. Lancet Neurol 12:264–274PubMedCentralCrossRefPubMed Wenning GK, Geser F, Krismer F, Seppi K, Duerr S, Boesch S, Kollensperger M, Goebel G, Pfeiffer KP, Barone P, Pellecchia MT, Quinn NP, Koukouni V, Fowler CJ, Schrag A, Mathias CJ, Giladi N, Gurevich T, Dupont E, Ostergaard K, Nilsson CF, Widner H, Oertel W, Eggert KM, Albanese A, del Sorbo F, Tolosa E, Cardozo A, Deuschl G, Hellriegel H, Klockgether T, Dodel R, Sampaio C, Coelho M, Djaldetti R, Melamed E, Gasser T, Kamm C, Meco G, Colosimo C, Rascol O, Meissner WG, Tison F, Poewe W, European Multiple System Atrophy Study Group (2013) The natural history of multiple system atrophy: a prospective European cohort study. Lancet Neurol 12:264–274PubMedCentralCrossRefPubMed
10.
Zurück zum Zitat Balreira A, Boczonadi V, Barca E, Pyle A, Bansagi B, Appleton M, Graham C, Hargreaves IP, Rasic VM, Lochmuller H, Griffin H, Taylor RW, Naini A, Chinnery PF, Hirano M, Quinzii CM, Horvath R (2014) ANO10 mutations cause ataxia and coenzyme Q(1)(0) deficiency. J Neurol 261:2192–2198PubMedCentralCrossRefPubMed Balreira A, Boczonadi V, Barca E, Pyle A, Bansagi B, Appleton M, Graham C, Hargreaves IP, Rasic VM, Lochmuller H, Griffin H, Taylor RW, Naini A, Chinnery PF, Hirano M, Quinzii CM, Horvath R (2014) ANO10 mutations cause ataxia and coenzyme Q(1)(0) deficiency. J Neurol 261:2192–2198PubMedCentralCrossRefPubMed
Metadaten
Titel
Frequency of rare recessive mutations in unexplained late onset cerebellar ataxia
verfasst von
M. J. Keogh
H. Steele
K. Douroudis
A. Pyle
J. Duff
R. Hussain
T. Smertenko
H. Griffin
M. Santibanez-Koref
R. Horvath
P. F. Chinnery
Publikationsdatum
01.08.2015
Verlag
Springer Berlin Heidelberg
Erschienen in
Journal of Neurology / Ausgabe 8/2015
Print ISSN: 0340-5354
Elektronische ISSN: 1432-1459
DOI
https://doi.org/10.1007/s00415-015-7772-x

Weitere Artikel der Ausgabe 8/2015

Journal of Neurology 8/2015 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Neu im Fachgebiet Neurologie

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.