Skip to main content
Erschienen in: BMC Neurology 1/2015

Open Access 01.12.2015 | Research article

Dysferlinopathy in Switzerland: clinical phenotypes and potential founder effects

verfasst von: Jens A. Petersen, Thierry Kuntzer, Dirk Fischer, Maja von der Hagen, Angela Huebner, Veronika Kana, Johannes A. Lobrinus, Wolfram Kress, Elisabeth J. Rushing, Michael Sinnreich, Hans H. Jung

Erschienen in: BMC Neurology | Ausgabe 1/2015

Abstract

Background

Dysferlin is reduced in patients with limb girdle muscular dystrophy type 2B, Miyoshi myopathy, distal anterior compartment myopathy, and in certain Ethnic clusters.

Methods

We evaluated clinical and genetic patient data from three different Swiss Neuromuscular Centers.

Results

Thirteen patients from 6 non-related families were included. Age of onset was 18.8 ± 4.3 years. In all patients, diallelic disease-causing mutations were identified in the DYSF gene. Nine patients from 3 non-related families from Central Switzerland carried the identical homozygous mutation, c.3031 + 2T>C. A possible founder effect was confirmed by haplotype analysis. Three patients from two different families carried the heterozygous mutation, c.1064_1065delAA. Two novel mutations were identified (c.2869C>T (p.Gln957Stop), c.5928G>A (p.Trp1976Stop)).

Conclusions

Our study confirms the phenotypic heterogeneity associated with DYSF mutations. Two mutations (c.3031 + 2T>C, c.1064_1065delAA) appear common in Switzerland. Haplotype analysis performed on one case (c. 3031 + 2T>C) suggested a possible founder effect.
Hinweise
Jens A. Petersen and Thierry Kuntzer contributed equally to this work.

Competing interests

Disclosure of conflict of interest: on behalf of all authors, the corresponding author states that there is no conflict of interest.

Authors’ contributions

WK carried out part of the molecular genetic studies and drafted the manuscript. MvdH and AH carried out the haplotype analysis. EJR, VK and AL carried out the neuropathological analyses. DF and MS participated in the design and the coordination of the study and helped to draft the manuscript. JAP, TK and HHJ conceived of the study, and participated in its design and coordination and helped to draft the manuscript. All authors read and approved the final manuscript. All patients gave their written informed consent for genetic analysis. The study was performed in accordance with the Declaration of Helsinki and approved by the local ethics committee (Kantonale Ethikkommission Zürich).

Authors’ information

Not applicable.

Background

Dysferlin (DYSF) is a transmembrane protein linked to sarcolemmal repair mechanisms. Autosomal recessive mutations in the DYSF gene cause muscular dystrophies (MD): a limb girdle MD, the so-called LGMD 2B, with onset in the proximal lower limbs [1], and two distal MD, one initially affecting the gastrocnemius muscle during early adulthood, Miyoshi myopathy (MM) [2], and the other involving muscles of the anterior compartment (DMAT) [3]. Disease severity ranges from asymptomatic mutation carriers with elevated serum CK levels [4] and exercise intolerance [5] to severe functional disability [6]. Muscle weakness with pelvic girdle involvement on clinical examination distinguishes LGMD from MM. Other common phenotypes include a “proximo-distal” phenotype, characterized by simultaneous distal and proximal weakness onset [6]. Of note, a recent MRI study suggests that all patients have radiographic evidence of proximo-distal muscle involvement, regardless of the clinical phenotype [7].
The DYSF gene maps to chromosome 2p12-p14, contains 55 exons and is transcribed as an 8.5 Kb major transcript mainly expressed in skeletal and cardiac muscles. The protein product is a 230 kDa molecule involved in muscle fibre repair [8, 9]. More than 400 DYSF mutations have been described in the Leiden muscular dystrophy database (http://​www.​dmd.​nl); however, a mutational hot spot has not been detected [1012]. Ethnic clusters have been described in Jews from Libya [13] and the Caucasus region [14], as well as in the Italian [15] and Spanish [16] populations. Four mutations account for 60 % of all mutations in Japanese patients with MM [17]. Interestingly, the type of mutation does not correlate with phenotypic features. In fact, the same mutation has been found to be associated with a broad spectrum of inter—and intra-familial clinical phenotypes [1822].
Within this range of clinical and genetic heterogeneity, we sought to determine frequently encountered DYSF gene mutations and phenotypes in the Swiss population and to uncover possible founder effects.

Methods

Subjects

The medical records of patients of Swiss origin with progressive muscle weakness, and DYSF mutations, who were admitted to Neuromuscular Centres in Basel, Lausanne and Zurich between 1989 and 2015 were anonymized and reviewed. All patients provided their written informed consent for genetic analysis. Clinical data collected included age and symptoms at onset, disease duration, family pedigrees, and muscle strength according to the Medical Research Council (MRC) scale. In addition, CK levels, muscle biopsy and mutational analysis results were evaluated. Electromyography, electrocardiography, echocardiography, pulmonary function and muscle MRI data were reviewed when available. The study was performed in accordance with the Declaration of Helsinki and approved by the local ethics committee (Kantonale Ethikkommission Zürich, KEK-ZH-Nr. 2015–0036).

Genetic analysis

Sequence analysis was performed using bidirectional fluorescent sequencing of all 55 exons of the DYSF gene, either in the Service de Cardiologie, Hôpital Cochin CHU Paris, France, or at the Institute of Human Genetics, University of Würzburg, Germany (Reference sequence: NM_003494.3). Multiplex ligation-dependent probe amplification (MLPA) was used in one case.
To evaluate the hypothesis that the DYSF mutation c.3031 + 2T>C is due to a founder effect, we performed haplotype analysis, as described previously, with 6 polymorphic microsatellite markers on chromosome 2p13.2 flanking the DYSF gene (D2S327, D2S2113, D2S292, D2S291, D21S2110 and D2S2111) [23].

Results

Clinical data

Thirteen patients were included in the study. Of those, 9 patients were from three non-related families from Central Switzerland (Canton of Schwyz). The clinical data of 10 patients from 6 different families are summarized in Tables 1 and 2. Three patients from central Switzerland (C-6, D-10, D-11) provided blood for haplotype analysis but refused clinical evaluation. Consanguinity was not reported and the parents of all patients were healthy. Mean disease onset was in late adolescence or early adulthood (18.7 ± 4.5 years). Early manifestations (Table 1) included leg pain, steppage gait, Gowers’ sign and difficulty running and jumping. Mean CK levels were 12500 ± 14400 IU/L (range 2200–46350, normal value for our laboratory < 167 U/l, Table 1).
Table 1
Clinical data of Swiss dysferlinopathy patients
Family-patient
Origin (Canton)
Sex/age (years)
Age at onset (years)
Symptoms at onset
CK level (IU/L)
Cardiac ultrasound/ECG
Pulmonary function
EMG
A-1
Fribourg
M/58
23
Steppage gait
>2.300
normal/normal
FVC 55 %
n.d.
A-2
 
F/54
28
Difficulty walking on tiptoes
>2.300
normal/normal
normal
Tibialis anterior muscle: myopathic, fibrillations
B-3
Aargau
F/33
15
Difficulty running
3.800
normal/normal
normal
Biceps brachii muscle: myopathic
C-4
Schwyz
M/27
17
Difficulty skiing
>23.000
normal/normal
normal
Vastus lateralis muscle:
    
(downhill) and running;
   
myopathic, fibrillations,
    
leg pain
   
positive sharp waves
C-5
 
F/24
17
Difficulty walking on
 >15.000
normal/normal
normal
Tibialis anterior muscle:
    
tiptoes; leg pain
   
myopathic
C-6
 
F/31
 
blood sample provided
    
    
for haplotype analysis;
    
    
consultation rejected
    
C-7
 
M/16
14
 Difficulty toe walking
 46.350
normal/normal
normal
Vastus lateralis muscle:
        
myopathic; positive
        
sharp waves
C-8
 
F/54
16
 Leg weakness
 n.d.
n.d.
n.d.
Extensor carpi radialis
        
muscle: myopathic
D-9
Schwyz
F/45
16
Gowers’ sign
>2.200
normal/normal
n.d.
Vastus lateralis muscle:
        
myopathic; fibrillations,
        
positive sharp waves
D-10
 
F/46
 
blood sample provided
    
    
for haplotype
    
D-11
 
M/48
 
analysis;
    
    
consultation rejected
    
E-12
Thurgau
M/28
20
Difficulty jumping,
>10.600
normal/normal
normal
n.d. (syncope during
    
Leg pain
   
needle insertion)
F-13
Schwyz
M/22
21
Leg weakness
>9.000
normal/normal
normal
Gastrocnemius muscle:
        
myopathic; fibrillations, positive sharp waves
M male, F female, FVC Forced vital capacity, n.d. not done, normal CK < 167 U/l. “myopathic” refers to small, short, polyphasic potentials
Table 2
Phenotype of Swiss dysferlinopathy patients
Family-patient
A-1
A-2
B-3
C-4
C-5
C-7
C-8
D-9
E-12
F-13
Age at onset
23
28
15
17
17
14
16
16
20
21
Age at examination/years after onset
46/23
55/32
45/17
52/24
30/15
20/3
27/11
18 /1
26/9
15/1
18/4
44/28
36/20
27/7
31/11
21/0
Facial muscles
5/5
5/5
nd
nd
5/5
5/5
5/5
5/5
5/5
5/5
5/5
5/5
5/5
5/5
5/5
5/5
Head extension
5/5
5/5
5/5
5/5
nd
5/5
5/5
5/5
5/5
5/5
5/5
4/4
nd
5/5
nd
5/5
Shoulder elevation
5/5
5/5
5/5
5/5
5/5
5/5
5/5
5/5
5/5
5/5
5/5
nd
nd
nd
nd
5/5
External arm rotation
3/3
0/0
4+/4+
5-/5-
nd
5/5
nd
nd
nd
5/5
nd
0/0
nd
nd
nd
nd
Internal arm rotation
3/3
nd
nd
nd
nd
5/5
nd
nd
nd
5/5
nd
nd
nd
nd
nd
nd
Arm adduction
3/3
0/0
nd
nd
3/3
5/5
nd
nd
nd
5/5
4/4
nd
nd
5/5
4+/4+
nd
Arm abduction
1/1
0/0
4+/4+
5/5
3/3
5/5
5-/5-
5/5
5/5
5/5
4/5
0/0
4/4
5/5
4-/4-
5/5
Arm flexion
nd
0/0
4+/4+
4/4
4/4
5/5
5-/5-
5/5
5/5
5/5
4/4
0/0
4/4
5/5
4-/4-
5/5
Arm extension
1/1
1/1
4+/4+
4+/4+
3/3
5/5
5-/5-
5/5
5/5
5/5
4+/4+
0/0
4/4
5/5
4/4
5/5
Wrist supination
4+/4
nd
nd
nd
nd
nd
nd
nd
5/5
5/5
nd
nd
nd
nd
nd
nd
Wrist pronation
4+/3
3/3
4+/4+
5/5
nd
nd
nd
nd
5/5
5/5
nd
nd
nd
nd
nd
nd
Hand flexion
nd
3+/3+
4+/4+
5/5
4/4
5/5
5-/5-
5/5
5/5
5/5
4/4
4+/4
5-/5-
5/5
4+/4+
5/5
Finger flexion
3/3
3-/3-
4+/4+
4/4
4/4
5/5
5-/5-
5/5
5/5
5/5
5/5
4+/4
5-/5-
5/5
4+/4+
5/5
Hand extension
5/5
4+/4
nd
nd
4/4
5/5
5-/5-
5/5
4/4
5/5
4/4
4+/4
5-/5-
5/5
4+/4+
5/5
Finger extension
5/5
4-/4-
5-/5-
5/5
4/4
5/5
5-/5-
5/5
4/4
5/5
4/4
4+/4
5-/5-
5/5
4+/4+
5/5
Thumb abduction
nd
4/4
4+/4+
4+/4+
nd
5/5
nd
nd
nd
5/5
nd
nd
nd
nd
nd
nd
Thumb extension
5/5
nd
nd
nd
nd
5/5
nd
nd
nd
5/5
nd
nd
nd
nd
nd
nd
Hip adduction
2/2
0/0
3/3
2+/2+
4/4
5/5
nd
5-/5-
nd
5/5
2/2
nd
4/4
nd
4+/4+
nd
Hip extension
5/5
4+/4+
4+/4+
4-/4-
nd
4/4
4-/4-
5-/5-
nd
5/5
4/4
1/1
4/4
nd
4-/4-
5/5
Knee extension
2/2
0/0
2+/2
2/2
2/2
5/5
4/4
5/5
5-/5-
5/5
4/4
3/3
3/3
5/5
4/4
5/5
Knee flexion
2/2
0/0
3+/3+
3-/3-
2/2
5/5
4-/4-
5/5
3+/3+
5/5
4/4
4/4
4/4
5/5
4-/4-
5/5
Foot extension
0/0
0/0
2/2
2/2
3/3
5/5
2/2
5/5
2/2
4/4
4/4
0/0
5-/5-
4/4
4/4
4/4
Toe extension
0/0
0/0
3-/3-
nd
nd
nd
nd
nd
nd
nd
nd
1/1
nd
3/3
nd
nd
Foot flexion
5/5
3/3
1/1
1/1
1/1
4-/4
2/1
5-/5-
3/3
4/4
3/3
1/1
4+/4+
4/4
4/4
3/4
Toe flexion
4+/4+
3/3
1/1
1/1
nd
nd
nd
nd
nd
nd
nd
4/4
nd
4-/4-
nd
nd
Muscle strength grades according to Medical Research Council (MRC) scale (right/left); nd: not done
The clinical phenotype could be determined in 10 patients (Table 2). In all patients, the facial muscles were spared. Four of the patients from Central Switzerland (C-4, C-5, C-7, F-13) presented without arm weakness a few years after the onset of symptoms (Table 2). All patients had distal lower limb flexor weakness and 2 (C-7, F-13) also showed distal lower limb extensor weakness. Three (C-4, C-5, C-7) patients had proximal lower limb weakness and therefore were classified as the “proximo-distal” (PD) phenotype [6]. Another patient (F-13) demonstrated isolated distal lower limb weakness (foot flexion > extension) and was classified as the MM phenotype. Three patients (C-4, C-5 and C-7) were followed for 11, 9 and 4 years, respectively (Table 2). All developed distal upper limb extensor weakness with 2 (DC-4, C-7) additionally showing distal upper limb flexor and proximal upper limb weakness. Two patients from central Switzerland (C-8, D-9) were examined 20 and 28 years after onset and demonstrated proximal and distal upper and lower limb flexor and extensor weakness (Table 2). Patient D-9 experienced a relatively “benign” course with muscle strength grade of at least 3 on the MRC scale (Table 2). One patient from the Canton Thurgau (E-12) presented 7 years after onset with proximal and distal lower limb flexor and extensor weakness, eventually developing distal flexor and extensor and proximal upper limb weakness (Table 2). A patient from the Canton Aargau (B-3) presented with severe distal lower limb flexor, proximal lower limb flexor and extensor, distal upper limb flexor and extensor and proximal upper limb weakness 15 years after onset. Two patients from the Canton Fribourg were initially examined 23 (A-1) and 17 (A-2) years after onset, respectively. Patient A-1 had generalized weakness, pronounced in the distal lower limb extensors, and over the course of 9 years developed proximal lower and upper limb palsies. Patient A-2 had generalized weakness, more pronounced in the distal lower limb flexors, that progressive minimally over 7 years. Interestingly, members of family C presented with a proximo-distal phenotype early in the course and members of family C and patient F-13 were notable for the lack of upper limb weakness during the early phases of the disease. Later in the course, all patients manifested a typical LGMD phenotype; however, the distribution of weakness was heterogeneous (Table 2). The DMAT phenotype was not observed in our cohort.
The use of mobility aids showed the following timeline: 2 patients required canes for walking assistance (A-1, A-2) at the age of 35 and 49 years, respectively (disease duration until first use of canes 12 and 21 years, respectively), and both were eventually wheelchair-bound (17 and 24 years after disease onset; 5 and 3 years after first cane use). Patient B-3, at age 29, 14 years after disease onset, used a wheelchair intermittently (B-3), and patient C-8 was wheelchair-bound, at age 43, 27 years after disease onset.

Cardiac, pulmonary, MRI and EMG data

Electrocardiography and ECG were normal in all 9 patients who underwent examination, the longest disease duration being 35 years (Table 1). Pulmonary function was normal in 6 out of 7 patients, with restrictive lung disease documented in patient A-1 (Table 1; initial pulmonary function testing performed after 29 years of disease duration). In 3 patients, muscle MRI showed fatty replacement of the lower limb muscles. In all three patients (A-2, E-12 and F-13), the short head of the biceps femoris and the sartorius and the gracilis muscles were symmetrically spared or appeared less affected than other muscles. In patients E-12 and F-13, the semitendinosus, the tibialis anterior and the extensor digitorum longus muscles also seemed less affected, while edema was suspected in selected lower limb muscles. In another patient (C-7), the pelvic and the lower limb muscles appeared normal. EMG was myopathic with short and small potentials in all 8 patients examined, with spontaneous activity found in 5 cases (Table 1).

Pathological features and protein studies

A total of 5 surgical muscle biopsies were available (Table 3) for evaluation. In 2 patients, biopsies were not available because they were taken in 1986 and 1977. In 6 patients, the genetic diagnosis was made without prior biopsy. The mean age at biopsy was 28.25 ± 13.7 years, while the mean disease duration at biopsy was 8.3 ± 9.8 years. Samples were removed from the trapezius, quadriceps and gracilis muscles. All patients exhibited dystrophic changes with an increased spectrum of myofiber diameter and variable fibrosis. In one case with a LGMD phenotype (B-3), striking inflammatory changes were observed within the endomysium. Western blots (WB) and/or immunohistochemistry (IHC) were performed in 6 samples (WB only in 2 samples, IHC only in 3 samples, WB and IHC in a single sample). On WB, dysferlin was totally absent in all samples tested, whereas by IHC, dysferlin was judged absent in 2 cases and reduced in two.
Table 3
Histological and biochemical data of Swiss dysferlinopathy patients
Family-patient
Disease duration at biopsy (y)/muscle/findings
WB Dysf/immunohistochemistry
A-1
26/trapezius/atrophic fibers; slight fibrosis; fatty infiltration
absent/nd
A-2
Biopsy in 1986; histopathological data na
absent/nd
B-3
1/vastus lateralis/atrophic fibers; fibre size variability;/re- and degenerating fibers; necrotic fibers; endo- and perimysial fibrosis; inflammation
absent/reduced
C-4
3/vastus lateralis/de- and regenerating fibers,
nd/absent
 
whorled fibers, myophagocytosis, endomysial fibrosis
 
C-5 to C-7
Genetic testing without prior biopsy
 
C-8
Biopsy in 1977; na
 
D-9
20/vastus lateralis/focal fiber atrophy, hypertrophy,
nd/reduced
 
necrotic fibers, increased internalized nuclei
 
D-10, D-11
Genetic testing without prior biopsy
 
E-12
1/gracilis/re- and degenerating fibers,
nd/absent
 
myophagocytosis, fibrosis, COX-neg. fibers
 
F-13
Genetic testing without prior biopsy
 
Nd not done, WB Dysf result of the presence or absence of dysferlin by Western-blot analysis

Genetic studies

Genetic data are summarized in Table 4. In 13 patients, 2 disease-causing mutations on separate alleles were identified. Nine patients from 3 non-related families from Central Switzerland (Canton Schwyz) carried the identical splice site mutation, c.3031 + 2T>C, 7 were homozygous (Table 4). Three patients from 2 different families from the Cantons of Fribourg and Aargau carried a heterozygous frameshift mutation c.1064_1065delAA, and 2 members from family A, harbored the heterozygous nonsense mutation c.2217C>A (Table 4).
Table 4
Dysferlin mutations in Swiss patients
Family-patient
Exon/Intron
DNA
Effect of mutation
A-1
ex12
c.1064_1065delAA
p.Lys355ArgfsX4
 
ex23
c.2217C>A
p.Tyr739X
A-2
ex12
c.1064_1065delAA
p.Lys355ArgfsX4
 
ex23
c.2217C>A
p.Tyr739X
B-3
ex12
c.1064/1065delAA
p.Lys355ArgfsX4
 
ex25-27
 
Deletion exon 25-27
C-4
IVS28
c. 3031 + 2T>C
Abn. Spli.
 
IVS28
c. 3031 + 2T>C
Abn. Spli.
C-5
IVS28
c. 3031 + 2T>C
Abn. Spli.
 
IVS28
c. 3031 + 2T>C
Abn. Spli.
C-6
IVS28
c. 3031 + 2T>C
Abn. Spli.
 
ex27
c. 2869C>T
p.Gln957X
C-7
IVS28
c. 3031 + 2T>C
Abn. Spli.
 
IVS28
c. 3031 + 2T>C
Abn. Spli.
C-8
IVS28
c.3031 + 2T>C
Abn. Spli
 
IVS28
c.3031 + 2T>C
Abn. Spli
D-9
IVS28
c. 3031 + 2T>C
Abn. Spli.
 
IVS28
c. 3031 + 2T>C
Abn. Spli.
D-10
IVS28
c. 3031 + 2T>C
Abn. Spli.
 
IVS28
c. 3031 + 2T>C
Abn. Spli.
D-11
IVS28
c. 3031 + 2T>C
Abn. Spli.
 
IVS28
c. 3031 + 2T>C
Abn. Spli.
E-13
Ex53
c.5928G>A
p.Trp1976Stop
 
Ex53
c.5928G>A
p.Trp1976Stop
F-13
IVS28
c. 3031 + 2T>C
Abn. Spli.
 
IVS28
c. 3031 + 2T>C
Abn. Spli.
Novel mutations appear in bold; Ex exon, IVS intervening sequence
Two novel mutations were identified in this patient set. Patient C-6, who was compound heterozygous for the c.3031 + 2T >C mutation on one allele, showed a novel mutation, c.2869C>T on the other allele. Patient E-12 carried the novel homozygous c.5928G > A mutation.
Haplotype analyses using markers flanking the DYSF gene revealed homozygosity for at least one nearest marker (D2S291) in all patients from the two apparently unrelated C and D families carrying the homozygous c.3031 + 2T>C mutation, suggesting a possible founder mutation (See Fig. 1). In patient C-7, carrying a compound heterozygous DYSF mutation (c.3031 + 2 T >C and c.2869C>T), marker D2S291 shows a heterozygous genotype (Fig. 1).

Discussion

Evaluation of the present cohort of 6 families brings new genetic information about dysferlinopathy in Switzerland, and novel data regarding a potential founder effect in Central Switzerland. The fact that unrelated families with identical mutations come from the same region indicates that these mutations are common, and indeed, haplotype analysis of 2 families carrying the mutation c.3031 + 2T>C suggests a possible founder effect for this mutation in Switzerland. The limitations of this study are twofold: the analysis is limited by the small cohort size and 3 out of 13 patients had not been examined clinically.
From a clinical point of view, the onset age in early adulthood confirms previous findings [6, 7, 24]. In addition, late-onset [25] or congenital forms were not encountered [26]. Three out of 4 patients from Central Switzerland presented with the PD phenotype, while another patient with the same mutation was considered to represent the MM phenotype. Upper limb weakness was not a presenting feature in the current cohort. Some reports suggest that the proximo-distal phenotype occurs in up to one third of dysferlinopathy cases [6]. Of note, 9 out of 10 patients developed distal upper limb weakness later in the disease course, which has been frequently reported [2729]. Muscle MRI is known to reveal fatty replacement of distal and proximal lower limb muscles in the early stages of dysferlinopathy, including patients with clinically isolated proximal or distal involvement [7, 30]. However, an adolescent patient with a MM phenotype (C-7) showed a normal pelvic and whole lower limb muscle MRI, similarly to a patient described with isolated hyperCKemia [31].
Interestingly, we found clinical phenotype variability within families and within unrelated carriers with similar mutations. Such variability has already been reported in Libyan Jews [13] and was found to be particularly relevant in a 6-generation, highly consanguineous family originating from an isolated village in Dagestan [32]. This observation may indicate that to date, unknown genetic or environmental modifiers play a role in influencing the clinical phenotypes of the mutated DYSF gene [22]. Accordingly, it would be interesting to study micro-RNAs known to be involved in many biological processes, including epigenetic changes. Other cardinal features of our cohort were very high CK levels, as previously described [7], the absence of cardiac impairment (nevertheless encountered in few patients [6, 25, 33, 34] and present in a knock out mouse model [35]), and the risk of developing respiratory failure late in the disease course [36, 37], as observed in one of our patients.
EMG showed myopathic changes and pathologic spontaneous activity. In one case, the biopsy revealed striking endomysial inflammation, in line with frequently observed inflammatory changes in dysferlinopathy and other muscular dystrophies [18, 38]. Immunosuppressive treatment, however, fails to improve muscle weakness [39]. It has been postulated that dysferlin may be linked to membrane repair and/or inflammatory activation [40], and in a few cases, splice site mutations disrupting dysferlin are known to produce inflammation [18]. Conversely, most of our patients with splice mutations showed no evidence of inflammation on muscle biopsy, indicating the lack of clear correlation between inflammation and genotype. On Western blot (WB) 3 cases showed a consistent reduction in dysferlin, as has been previously described [41].
In dysferlinopathies, genotype is not predictive of disease severity, and genotype-phenotype correlations are not clear-cut [15, 42]. However, correlation has been found between certain geographical areas and the age of onset, notably in Japanese and Italian MM patients [17], who develop symptoms earlier than patients with LGMD 2B [10]. The most severely affected patients in our study (Patients A-1 and A-2, described previously [12]) carried the truncating mutation c.2217C>A (p.Tyr739X) with a disease duration of 17 and 24 years, respectively, before the patients became wheelchair-bound. One of the older patients (C-8), carrying the c.3031 + 2T>C homozygous mutation, became wheelchair bound after a disease duration of 27 years, and patient D-9 showed relatively mild impairment 20 years after onset. However, in all other patients carrying this mutation, disease duration was shorter; thus precluding longitudinal comparisons. A longer follow-up is necessary to assess the onset and time-course of the various disabling symptoms.

Conclusion

Our study confirms the broad phenotypic heterogeneity, often accompanied by markedly elevated CK levels, associated with DYSF mutations in the Swiss population. Due to a potential founder effect in the past, two mutations appear to be endemic in central Switzerlans. These findings are important for genetic counselling and should facilitate targeted molecular diagnosis of dysferlin deficiency in patients of Swiss origin.

Acknowledgments

We thank P. Mitzscherling for expert technical assistance.
MvdH, AH and WK are members of the German network on muscular dystrophies (MD-NET, 01GM0601) which is funded by the German Ministry of Education and Research (BMBF, Bonn, Germany).
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. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

Disclosure of conflict of interest: on behalf of all authors, the corresponding author states that there is no conflict of interest.

Authors’ contributions

WK carried out part of the molecular genetic studies and drafted the manuscript. MvdH and AH carried out the haplotype analysis. EJR, VK and AL carried out the neuropathological analyses. DF and MS participated in the design and the coordination of the study and helped to draft the manuscript. JAP, TK and HHJ conceived of the study, and participated in its design and coordination and helped to draft the manuscript. All authors read and approved the final manuscript. All patients gave their written informed consent for genetic analysis. The study was performed in accordance with the Declaration of Helsinki and approved by the local ethics committee (Kantonale Ethikkommission Zürich).

Authors’ information

Not applicable.
Literatur
1.
Zurück zum Zitat Bashir R, Britton S, Strachan T, Keers S, Vafiadaki E, Lako M, et al. A gene related to Caenorhabditis elegans spermatogenesis factor fer-1 is mutated in limb-girdle muscular dystrophy type 2B. Nat Genet. 1998;20(1):37–42.CrossRefPubMed Bashir R, Britton S, Strachan T, Keers S, Vafiadaki E, Lako M, et al. A gene related to Caenorhabditis elegans spermatogenesis factor fer-1 is mutated in limb-girdle muscular dystrophy type 2B. Nat Genet. 1998;20(1):37–42.CrossRefPubMed
2.
Zurück zum Zitat Liu J, Aoki M, Illa I, Wu C, Fardeau M, Angelini C, et al. Dysferlin, a novel skeletal muscle gene, is mutated in Miyoshi myopathy and limb girdle muscular dystrophy. Nat Genet. 1998;20(1):31–6.CrossRefPubMed Liu J, Aoki M, Illa I, Wu C, Fardeau M, Angelini C, et al. Dysferlin, a novel skeletal muscle gene, is mutated in Miyoshi myopathy and limb girdle muscular dystrophy. Nat Genet. 1998;20(1):31–6.CrossRefPubMed
3.
Zurück zum Zitat Illa I, Serrano-Munuera C, Gallardo E, Lasa A, Rojas-Garcia R, Palmer J, et al. Distal anterior compartment myopathy: a dysferlin mutation causing a new muscular dystrophy phenotype. Ann Neurol. 2001;49(1):130–4.CrossRefPubMed Illa I, Serrano-Munuera C, Gallardo E, Lasa A, Rojas-Garcia R, Palmer J, et al. Distal anterior compartment myopathy: a dysferlin mutation causing a new muscular dystrophy phenotype. Ann Neurol. 2001;49(1):130–4.CrossRefPubMed
4.
Zurück zum Zitat Fanin M, Nascimbeni AC, Aurino S, Tasca E, Pegoraro E, Nigro V, et al. Frequency of LGMD gene mutations in Italian patients with distinct clinical phenotypes. Neurology. 2009;72(16):1432–5.CrossRefPubMed Fanin M, Nascimbeni AC, Aurino S, Tasca E, Pegoraro E, Nigro V, et al. Frequency of LGMD gene mutations in Italian patients with distinct clinical phenotypes. Neurology. 2009;72(16):1432–5.CrossRefPubMed
5.
Zurück zum Zitat Xi J, Blandin G, Lu J, Luo S, Zhu W, Beroud C, et al. Clinical heterogeneity and a high proportion of novel mutations in a Chinese cohort of patients with dysferlinopathy. Neurol India. 2014;62(6):635–9.CrossRefPubMed Xi J, Blandin G, Lu J, Luo S, Zhu W, Beroud C, et al. Clinical heterogeneity and a high proportion of novel mutations in a Chinese cohort of patients with dysferlinopathy. Neurol India. 2014;62(6):635–9.CrossRefPubMed
6.
Zurück zum Zitat Nguyen K, Bassez G, Krahn M, Bernard R, Laforet P, Labelle V, et al. Phenotypic study in 40 patients with dysferlin gene mutations: high frequency of atypical phenotypes. Arch Neurol. 2007;64(8):1176–82.CrossRefPubMed Nguyen K, Bassez G, Krahn M, Bernard R, Laforet P, Labelle V, et al. Phenotypic study in 40 patients with dysferlin gene mutations: high frequency of atypical phenotypes. Arch Neurol. 2007;64(8):1176–82.CrossRefPubMed
7.
Zurück zum Zitat Paradas C, Llauger J, Diaz-Manera J, Rojas-Garcia R, De Luna N, Iturriaga C, et al. Redefining dysferlinopathy phenotypes based on clinical findings and muscle imaging studies. Neurology. 2010;75(4):316–23.CrossRefPubMed Paradas C, Llauger J, Diaz-Manera J, Rojas-Garcia R, De Luna N, Iturriaga C, et al. Redefining dysferlinopathy phenotypes based on clinical findings and muscle imaging studies. Neurology. 2010;75(4):316–23.CrossRefPubMed
8.
Zurück zum Zitat Lennon NJ, Kho A, Bacskai BJ, Perlmutter SL, Hyman BT, Brown RH. Dysferlin interacts with Annexins A1 and A2 and mediates sarcolemmal wound-healing. J Biol Chem. 2003;278(50):50466–73.CrossRefPubMed Lennon NJ, Kho A, Bacskai BJ, Perlmutter SL, Hyman BT, Brown RH. Dysferlin interacts with Annexins A1 and A2 and mediates sarcolemmal wound-healing. J Biol Chem. 2003;278(50):50466–73.CrossRefPubMed
9.
Zurück zum Zitat Bansal D, Miyake K, Vogel SS, Groh S, Chen CC, Williamson R, et al. Defective membrane repair in dysferlin-deficient muscular dystrophy. Nature. 2003;423(6936):168–72.CrossRefPubMed Bansal D, Miyake K, Vogel SS, Groh S, Chen CC, Williamson R, et al. Defective membrane repair in dysferlin-deficient muscular dystrophy. Nature. 2003;423(6936):168–72.CrossRefPubMed
10.
Zurück zum Zitat Cagliani R, Magri F, Toscano A, Merlini L, Fortunato F, Lamperti C, et al. Mutation finding in patients with dysferlin deficiency and role of the dysferlin interacting proteins annexin A1 and A2 in muscular dystrophies. Hum Mutat. 2005;26(3):283.CrossRefPubMed Cagliani R, Magri F, Toscano A, Merlini L, Fortunato F, Lamperti C, et al. Mutation finding in patients with dysferlin deficiency and role of the dysferlin interacting proteins annexin A1 and A2 in muscular dystrophies. Hum Mutat. 2005;26(3):283.CrossRefPubMed
11.
Zurück zum Zitat Nguyen K, Bassez G, Bernard R, Krahn M, Labelle V, Figarella-Branger D, et al. Dysferlin mutations in LGMD2B, Miyoshi myopathy, and atypical dysferlinopathies. Hum Mutat. 2005;26(2):165.CrossRefPubMed Nguyen K, Bassez G, Bernard R, Krahn M, Labelle V, Figarella-Branger D, et al. Dysferlin mutations in LGMD2B, Miyoshi myopathy, and atypical dysferlinopathies. Hum Mutat. 2005;26(2):165.CrossRefPubMed
12.
Zurück zum Zitat Krahn M, Beroud C, Labelle V, Nguyen K, Bernard R, Bassez G, et al. Analysis of the DYSF mutational spectrum in a large cohort of patients. Hum Mutat. 2009;30(2):E345–375.CrossRefPubMed Krahn M, Beroud C, Labelle V, Nguyen K, Bernard R, Bassez G, et al. Analysis of the DYSF mutational spectrum in a large cohort of patients. Hum Mutat. 2009;30(2):E345–375.CrossRefPubMed
13.
Zurück zum Zitat Argov Z, Sadeh M, Mazor K, Soffer D, Kahana E, Eisenberg I, et al. Muscular dystrophy due to dysferlin deficiency in Libyan Jews. Clinical and genetic features. Brain. 2000;123(Pt 6):1229–37.CrossRefPubMed Argov Z, Sadeh M, Mazor K, Soffer D, Kahana E, Eisenberg I, et al. Muscular dystrophy due to dysferlin deficiency in Libyan Jews. Clinical and genetic features. Brain. 2000;123(Pt 6):1229–37.CrossRefPubMed
14.
Zurück zum Zitat Leshinsky-Silver E, Argov Z, Rozenboim L, Cohen S, Tzofi Z, Cohen Y, et al. Dysferlinopathy in the Jews of the Caucasus: a frequent mutation in the dysferlin gene. Neuromuscul Disord. 2007;17(11–12):950–4.CrossRefPubMed Leshinsky-Silver E, Argov Z, Rozenboim L, Cohen S, Tzofi Z, Cohen Y, et al. Dysferlinopathy in the Jews of the Caucasus: a frequent mutation in the dysferlin gene. Neuromuscul Disord. 2007;17(11–12):950–4.CrossRefPubMed
15.
Zurück zum Zitat Cagliani R, Fortunato F, Giorda R, Rodolico C, Bonaglia MC, Sironi M, et al. Molecular analysis of LGMD-2B and MM patients: identification of novel DYSF mutations and possible founder effect in the Italian population. Neuromuscul Disord. 2003;13(10):788–95.CrossRefPubMed Cagliani R, Fortunato F, Giorda R, Rodolico C, Bonaglia MC, Sironi M, et al. Molecular analysis of LGMD-2B and MM patients: identification of novel DYSF mutations and possible founder effect in the Italian population. Neuromuscul Disord. 2003;13(10):788–95.CrossRefPubMed
16.
Zurück zum Zitat Vilchez JJ, Gallano P, Gallardo E, Lasa A, Rojas-Garcia R, Freixas A, et al. Identification of a novel founder mutation in the DYSF gene causing clinical variability in the Spanish population. Arch Neurol. 2005;62(8):1256–9.CrossRefPubMed Vilchez JJ, Gallano P, Gallardo E, Lasa A, Rojas-Garcia R, Freixas A, et al. Identification of a novel founder mutation in the DYSF gene causing clinical variability in the Spanish population. Arch Neurol. 2005;62(8):1256–9.CrossRefPubMed
17.
Zurück zum Zitat Takahashi T, Aoki M, Tateyama M, Kondo E, Mizuno T, Onodera Y, et al. Dysferlin mutations in Japanese Miyoshi myopathy: relationship to phenotype. Neurology. 2003;60(11):1799–804.CrossRefPubMed Takahashi T, Aoki M, Tateyama M, Kondo E, Mizuno T, Onodera Y, et al. Dysferlin mutations in Japanese Miyoshi myopathy: relationship to phenotype. Neurology. 2003;60(11):1799–804.CrossRefPubMed
18.
Zurück zum Zitat McNally EM, Ly CT, Rosenmann H, Mitrani Rosenbaum S, Jiang W, Anderson LV, et al. Splicing mutation in dysferlin produces limb-girdle muscular dystrophy with inflammation. Am J Med Genet. 2000;91(4):305–12.CrossRefPubMed McNally EM, Ly CT, Rosenmann H, Mitrani Rosenbaum S, Jiang W, Anderson LV, et al. Splicing mutation in dysferlin produces limb-girdle muscular dystrophy with inflammation. Am J Med Genet. 2000;91(4):305–12.CrossRefPubMed
19.
Zurück zum Zitat Aoki M, Liu J, Richard I, Bashir R, Britton S, Keers SM, et al. Genomic organization of the dysferlin gene and novel mutations in Miyoshi myopathy. Neurology. 2001;57(2):271–8.CrossRefPubMed Aoki M, Liu J, Richard I, Bashir R, Britton S, Keers SM, et al. Genomic organization of the dysferlin gene and novel mutations in Miyoshi myopathy. Neurology. 2001;57(2):271–8.CrossRefPubMed
20.
Zurück zum Zitat Nakagawa M, Matsuzaki T, Suehara M, Kanzato N, Takashima H, Higuchi I, et al. Phenotypic variation in a large Japanese family with Miyoshi myopathy with nonsense mutation in exon 19 of dysferlin gene. J Neurol Sci. 2001;184(1):15–9.CrossRefPubMed Nakagawa M, Matsuzaki T, Suehara M, Kanzato N, Takashima H, Higuchi I, et al. Phenotypic variation in a large Japanese family with Miyoshi myopathy with nonsense mutation in exon 19 of dysferlin gene. J Neurol Sci. 2001;184(1):15–9.CrossRefPubMed
21.
Zurück zum Zitat Ueyama H, Kumamoto T, Nagao S, Masuda T, Horinouchi H, Fujimoto S, et al. A new dysferlin gene mutation in two Japanese families with limb-girdle muscular dystrophy 2B and Miyoshi myopathy. Neuromuscul Disord. 2001;11(2):139–45.CrossRefPubMed Ueyama H, Kumamoto T, Nagao S, Masuda T, Horinouchi H, Fujimoto S, et al. A new dysferlin gene mutation in two Japanese families with limb-girdle muscular dystrophy 2B and Miyoshi myopathy. Neuromuscul Disord. 2001;11(2):139–45.CrossRefPubMed
22.
Zurück zum Zitat Weiler T, Bashir R, Anderson LV, Davison K, Moss JA, Britton S, et al. Identical mutation in patients with limb girdle muscular dystrophy type 2B or Miyoshi myopathy suggests a role for modifier gene(s). Hum Mol Genet. 1999;8(5):871–7.CrossRefPubMed Weiler T, Bashir R, Anderson LV, Davison K, Moss JA, Britton S, et al. Identical mutation in patients with limb girdle muscular dystrophy type 2B or Miyoshi myopathy suggests a role for modifier gene(s). Hum Mol Genet. 1999;8(5):871–7.CrossRefPubMed
23.
Zurück zum Zitat von der Hagen M, Schallner J, Kaindl AM, Koehler K, Mitzscherling P, Abicht A, et al. Facing the genetic heterogeneity in neuromuscular disorders: linkage analysis as an economic diagnostic approach towards the molecular diagnosis. Neuromuscul Disord. 2006;16(1):4–13.CrossRefPubMed von der Hagen M, Schallner J, Kaindl AM, Koehler K, Mitzscherling P, Abicht A, et al. Facing the genetic heterogeneity in neuromuscular disorders: linkage analysis as an economic diagnostic approach towards the molecular diagnosis. Neuromuscul Disord. 2006;16(1):4–13.CrossRefPubMed
24.
Zurück zum Zitat Nalini A, Gayathri N. Dysferlinopathy: a clinical and histopathological study of 28 patients from India. Neurol India. 2008;56(3):379–85. discussion 386–377.CrossRefPubMed Nalini A, Gayathri N. Dysferlinopathy: a clinical and histopathological study of 28 patients from India. Neurol India. 2008;56(3):379–85. discussion 386–377.CrossRefPubMed
25.
Zurück zum Zitat Klinge L, Aboumousa A, Eagle M, Hudson J, Sarkozy A, Vita G, et al. New aspects on patients affected by dysferlin deficient muscular dystrophy. J Neurol Neurosurg Psychiatry. 2010;81(9):946–53.CrossRefPubMed Klinge L, Aboumousa A, Eagle M, Hudson J, Sarkozy A, Vita G, et al. New aspects on patients affected by dysferlin deficient muscular dystrophy. J Neurol Neurosurg Psychiatry. 2010;81(9):946–53.CrossRefPubMed
26.
Zurück zum Zitat Paradas C, Gonzalez-Quereda L, De Luna N, Gallardo E, Garcia-Consuegra I, Gomez H, et al. A new phenotype of dysferlinopathy with congenital onset. Neuromuscul Disord. 2009;19(1):21–5.CrossRefPubMed Paradas C, Gonzalez-Quereda L, De Luna N, Gallardo E, Garcia-Consuegra I, Gomez H, et al. A new phenotype of dysferlinopathy with congenital onset. Neuromuscul Disord. 2009;19(1):21–5.CrossRefPubMed
27.
Zurück zum Zitat Linssen WH, Notermans NC, Van der Graaf Y, Wokke JH, Van Doorn PA, Howeler CJ, et al. Miyoshi-type distal muscular dystrophy. Clinical spectrum in 24 Dutch patients. Brain. 1997;120(Pt 11):1989–96.CrossRefPubMed Linssen WH, Notermans NC, Van der Graaf Y, Wokke JH, Van Doorn PA, Howeler CJ, et al. Miyoshi-type distal muscular dystrophy. Clinical spectrum in 24 Dutch patients. Brain. 1997;120(Pt 11):1989–96.CrossRefPubMed
28.
Zurück zum Zitat Rosales XQ, Gastier-Foster JM, Lewis S, Vinod M, Thrush DL, Astbury C, et al. Novel diagnostic features of dysferlinopathies. Muscle Nerve. 2010;42(1):14–21.CrossRefPubMedPubMedCentral Rosales XQ, Gastier-Foster JM, Lewis S, Vinod M, Thrush DL, Astbury C, et al. Novel diagnostic features of dysferlinopathies. Muscle Nerve. 2010;42(1):14–21.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Woudt L, Di Capua GA, Krahn M, Castiglioni C, Hughes R, Campero M, Trangulao A, Gonzalez-Hormazabal P, Godoy-Herrera R, Levy N et al.: Toward an objective measure of functional disability in dysferlinopathy. Muscle Nerve 2015 doi:10.1002/mus.24685 Woudt L, Di Capua GA, Krahn M, Castiglioni C, Hughes R, Campero M, Trangulao A, Gonzalez-Hormazabal P, Godoy-Herrera R, Levy N et al.: Toward an objective measure of functional disability in dysferlinopathy. Muscle Nerve 2015 doi:10.​1002/​mus.​24685
30.
Zurück zum Zitat Fischer D, Walter MC, Kesper K, Petersen JA, Aurino S, Nigro V, et al. Diagnostic value of muscle MRI in differentiating LGMD2I from other LGMDs. J Neurol. 2005;252(5):538–47.CrossRefPubMed Fischer D, Walter MC, Kesper K, Petersen JA, Aurino S, Nigro V, et al. Diagnostic value of muscle MRI in differentiating LGMD2I from other LGMDs. J Neurol. 2005;252(5):538–47.CrossRefPubMed
31.
Zurück zum Zitat Kesper K, Kornblum C, Reimann J, Lutterbey G, Schroder R, Wattjes MP. Pattern of skeletal muscle involvement in primary dysferlinopathies: a whole-body 3.0-T magnetic resonance imaging study. Acta Neurol Scand. 2009;120(2):111–8.CrossRefPubMed Kesper K, Kornblum C, Reimann J, Lutterbey G, Schroder R, Wattjes MP. Pattern of skeletal muscle involvement in primary dysferlinopathies: a whole-body 3.0-T magnetic resonance imaging study. Acta Neurol Scand. 2009;120(2):111–8.CrossRefPubMed
32.
Zurück zum Zitat Illarioshkin SN, Ivanova-Smolenskaya IA, Tanaka H, Vereshchagin NV, Markova ED, Poleshchuk VV, et al. Clinical and molecular analysis of a large family with three distinct phenotypes of progressive muscular dystrophy. Brain. 1996;119(Pt 6):1895–909.CrossRefPubMed Illarioshkin SN, Ivanova-Smolenskaya IA, Tanaka H, Vereshchagin NV, Markova ED, Poleshchuk VV, et al. Clinical and molecular analysis of a large family with three distinct phenotypes of progressive muscular dystrophy. Brain. 1996;119(Pt 6):1895–909.CrossRefPubMed
33.
Zurück zum Zitat Wenzel K, Geier C, Qadri F, Hubner N, Schulz H, Erdmann B, et al. Dysfunction of dysferlin-deficient hearts. J Mol Med. 2007;85(11):1203–14.CrossRefPubMed Wenzel K, Geier C, Qadri F, Hubner N, Schulz H, Erdmann B, et al. Dysfunction of dysferlin-deficient hearts. J Mol Med. 2007;85(11):1203–14.CrossRefPubMed
34.
Zurück zum Zitat Ueyama H, Kumamoto T, Horinouchi H, Fujimoto S, Aono H, Tsuda T. Clinical heterogeneity in dysferlinopathy. Intern Med. 2002;41(7):532–6.CrossRefPubMed Ueyama H, Kumamoto T, Horinouchi H, Fujimoto S, Aono H, Tsuda T. Clinical heterogeneity in dysferlinopathy. Intern Med. 2002;41(7):532–6.CrossRefPubMed
35.
Zurück zum Zitat Han R, Bansal D, Miyake K, Muniz VP, Weiss RM, McNeil PL, et al. Dysferlin-mediated membrane repair protects the heart from stress-induced left ventricular injury. J Clin Invest. 2007;117(7):1805–13.CrossRefPubMedPubMedCentral Han R, Bansal D, Miyake K, Muniz VP, Weiss RM, McNeil PL, et al. Dysferlin-mediated membrane repair protects the heart from stress-induced left ventricular injury. J Clin Invest. 2007;117(7):1805–13.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Takahashi T, Aoki M, Suzuki N, Tateyama M, Yaginuma C, Sato H, et al. Clinical features and a mutation with late onset of limb girdle muscular dystrophy 2B. J Neurol Neurosurg Psychiatry. 2013;84(4):433–40.CrossRefPubMed Takahashi T, Aoki M, Suzuki N, Tateyama M, Yaginuma C, Sato H, et al. Clinical features and a mutation with late onset of limb girdle muscular dystrophy 2B. J Neurol Neurosurg Psychiatry. 2013;84(4):433–40.CrossRefPubMed
37.
Zurück zum Zitat Mahjneh I, Marconi G, Bushby K, Anderson LV, Tolvanen-Mahjneh H, Somer H. Dysferlinopathy (LGMD2B): a 23-year follow-up study of 10 patients homozygous for the same frameshifting dysferlin mutations. Neuromuscul Disord. 2001;11(1):20–6.CrossRefPubMed Mahjneh I, Marconi G, Bushby K, Anderson LV, Tolvanen-Mahjneh H, Somer H. Dysferlinopathy (LGMD2B): a 23-year follow-up study of 10 patients homozygous for the same frameshifting dysferlin mutations. Neuromuscul Disord. 2001;11(1):20–6.CrossRefPubMed
38.
Zurück zum Zitat Fanin M, Angelini C. Muscle pathology in dysferlin deficiency. Neuropathol Appl Neurobiol. 2002;28(6):461–70.CrossRefPubMed Fanin M, Angelini C. Muscle pathology in dysferlin deficiency. Neuropathol Appl Neurobiol. 2002;28(6):461–70.CrossRefPubMed
39.
Zurück zum Zitat Walter MC, Reilich P, Thiele S, Schessl J, Schreiber H, Reiners K, et al. Treatment of dysferlinopathy with deflazacort: a double-blind, placebo-controlled clinical trial. Orphanet J Rare Dis. 2013;8:26.CrossRefPubMedPubMedCentral Walter MC, Reilich P, Thiele S, Schessl J, Schreiber H, Reiners K, et al. Treatment of dysferlinopathy with deflazacort: a double-blind, placebo-controlled clinical trial. Orphanet J Rare Dis. 2013;8:26.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Cacciottolo M, Numitone G, Aurino S, Caserta IR, Fanin M, Politano L, et al. Muscular dystrophy with marked Dysferlin deficiency is consistently caused by primary dysferlin gene mutations. Eur J Hum Genet. 2011;19(9):974–80.CrossRefPubMedPubMedCentral Cacciottolo M, Numitone G, Aurino S, Caserta IR, Fanin M, Politano L, et al. Muscular dystrophy with marked Dysferlin deficiency is consistently caused by primary dysferlin gene mutations. Eur J Hum Genet. 2011;19(9):974–80.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Kawabe K, Goto K, Nishino I, Angelini C, Hayashi YK. Dysferlin mutation analysis in a group of Italian patients with limb-girdle muscular dystrophy and Miyoshi myopathy. Eur J Neurol. 2004;11(10):657–61.CrossRefPubMed Kawabe K, Goto K, Nishino I, Angelini C, Hayashi YK. Dysferlin mutation analysis in a group of Italian patients with limb-girdle muscular dystrophy and Miyoshi myopathy. Eur J Neurol. 2004;11(10):657–61.CrossRefPubMed
Metadaten
Titel
Dysferlinopathy in Switzerland: clinical phenotypes and potential founder effects
verfasst von
Jens A. Petersen
Thierry Kuntzer
Dirk Fischer
Maja von der Hagen
Angela Huebner
Veronika Kana
Johannes A. Lobrinus
Wolfram Kress
Elisabeth J. Rushing
Michael Sinnreich
Hans H. Jung
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
BMC Neurology / Ausgabe 1/2015
Elektronische ISSN: 1471-2377
DOI
https://doi.org/10.1186/s12883-015-0449-3

Weitere Artikel der Ausgabe 1/2015

BMC Neurology 1/2015 Zur Ausgabe