The congenital myopathies are a genetically hetero-
geneous group of early-onset neuromuscular disorders
characterized by variable degrees of muscle weakness
and distinctive structural abnormalities in muscle biopsy
samples. The conditions that have been identified to date
are mostly disorders of muscle excitation–contraction
coupling (ECC) or of proteins primarily involved in
sarcomeric filament assembly and interaction. However,
recent findings suggest other less common pathogenic
mechanisms. The concept of congenital myopathies was
established in the 1950s and 1960s, when the application
of histochemical and ultrastructural techniques to dis-
eased muscle identified histopathological features that
were considered to be pathognomonic. Recognition of
these features — namely, central cores, multi-minicores,
central nuclei and nemaline rods — resulted in the desig-
nation of four novel disease entities, central core disease
(CCD)
1
, multi-minicore disease (MmD)
2
, centronuclear
myopathy (CNM)
3
and nemaline myopathy
4
, which still
represent the main diagnostic categories.
Considerable progress has been made in under-
standing the phenotypic spectrum, diagnosis and
management of the congenital myopathies. In addition
to primary myopathic features, non-neuromuscular
mani festations are observed in several forms, point-
ing to a role for the defective proteins in non-skeletal
muscle tissues
5
. Muscle imaging, in particular, muscle
MRI, has emerged as a powerful tool for deep pheno-
typing
6
. Presentations late in adulthood have now been
recognized
7,8
, and owing to improved standards of care,
even patients with severe early-onset forms increasingly
transition from paediatric to adult neurology services.
Since the identification of dominant mutations in
the skeletal muscle ryanodine receptor 1 (RYR1) gene
as the cause of malignant hyperthermia in 1991 and
CCD in 1993
9,10
, mutations in more than 20 genes have
been identified in patients with congenital myopathies.
Introduction of next-generation sequencing (NGS)
techniques into routine clinical diagnosis
11
has resulted
in an improved detection rate for mutations in genes
such as RYR1, nebulin (NEB) and titin (TTN). Owing to
their large size, these genes were previously only stud-
ied by Sanger sequencing in a few patients. Novel NGS
techniques have led to the recognition that different
mutations in the same gene can give rise to various histo-
pathological phenotypes, and that mutations in different
*e-mail: f.munt[email protected]
doi:10.1038/nrneurol.2017.191
Published online 2 Feb 2018
Congenital myopathies: disorders
ofexcitation–contraction coupling
andmuscle contraction
Heinz Jungbluth
1,2,3
, Susan Treves
4,5
, Francesco Zorzato
4,5
, Anna Sarkozy
6
, JulienOchala
7
,
Caroline Sewry
6
, Rahul Phadke
6
, Mathias Gautel
2
and FrancescoMuntoni
6,8
*
Abstract
|
The congenital myopathies are a group of early-onset, non-dystrophic neuromuscular
conditions with characteristic muscle biopsy findings, variable severity and a stable or slowly
progressive course. Pronounced weakness in axial and proximal muscle groups is a common
feature, and involvement of extraocular, cardiorespiratory and/or distal muscles can implicate
specific genetic defects. Central core disease (CCD), multi-minicore disease (MmD),
centronuclear myopathy (CNM) and nemaline myopathy were among the first congenital
myopathies to be reported, and they still represent the main diagnostic categories. However,
these entities seem to belong to a much wider phenotypic spectrum. To date, congenital
myopathies have been attributed to mutations in over 20 genes, which encode proteins
implicated in skeletal muscle Ca
2+
homeostasis, excitation–contraction coupling, thin–thick
filament assembly and interactions, and other mechanisms. RYR1 mutations are the most
frequent genetic cause, and CCD and MmD are the most common subgroups. Next-generation
sequencing has vastly improved mutation detection and has enabled the identification of novel
genetic backgrounds. At present, management of congenital myopathies is largely supportive,
although new therapeutic approaches are reaching the clinical trial stage.
REVIEWS
NATURE REVIEWS
|
NEUROLOGY VOLUME 14
|
MARCH 2018
|
151
genes can cause the same histopathological feature, often
owing to functional associations between the defective
proteins. Moreover, it has become increasingly clear that
many congenital myopathies are characterized by non-
specific or complex pathological abnormalities rather
than a ‘pure’ muscle pathology picture. A classification
based on predominant histopathological and associated
clinical features is still useful; however, it is also help-
ful to consider these conditions according to the main
underlying disease mechanisms.
In this Review, we summarize genetic, clinical and
pathological features of the main congenital myopathies.
Common pathogenic mechanisms, diagnostic and cur-
rent management approaches, and principles of therapy
development will be outlined.
Classification and epidemiology
Data concerning the precise epidemiology of the con-
genital myopathies are limited and are mostly focused on
the four main pathological variants: CCD, MmD, CNM
and nemaline myopathy. The key characteristics of these
entities are detailed below and illustrated in FIG.1.
CCD — initially described in the 1950s
1
— and
MmD
2
are often referred to as the ‘core myopathies
12
,
and their names are derived from the histochemical
appearance of focally reduced oxidative enzyme activ-
ity, which corresponds to myofibrillar changes on
ultrastructural examination. CCD is characterized by
centrally located, well-demarcated cores that run along
the fibre axis for a substantial distance on longitudinal
sections, whereas MmD is defined by multiple cores of
less well-defined appearance and more-limitedlength.
The hallmark of CNM is the presence of fibres with
centralized nuclei, which show variations in terms of
numbers and associated features between muscles and
genetic backgrounds. Nemaline myopathy is character-
ized by the presence of numerous nemaline rods that
stain red with Gomori trichrome and can be confirmed
by electron microscopy
13
.
The overall prevalence of these congenital myopathy
variants has been estimated at 1 in 26,000
14
. Nemaline
myopathy was originally considered to be the most fre-
quent form, but emerging data suggest that congenital
myopathies with cores (CCD and MmD) represent the
most common subgroup. Marked genetic heterogeneity
is now acknowledged, as detailed below. RYR1 seems to
be the gene most frequently involved in congenital myo-
pathies, in particular, CCD and MmD. Recessive NEB
mutations and denovo dominant mutations in ACTA1,
which encodes skeletal muscle α-actin, are the most
common known causes of nemaline myopathy, whereas
X-linked recessive mutations in the myotubularin gene
(MTM1) are believed to be the most common cause of
CNM. Mutations in TTN are increasingly recognized and
may be involved in a substantial proportion of currently
unresolved congenital myopathies as well as other neuro-
muscular disorders, including muscular dystrophies
15
.
The genes implicated in the congenital myo pathies are
listed in TABLE1, and the key clinicopathological fea-
tures associated with the most common genetic back-
grounds are summarized in TABLE2. Characteristic
histopathological features are illustrated in FIG.1.
Clinicopathology and genetics
Congenital myopathies with cores
In view of their pathological and genetic overlap, CCD,
MmD and malignant hyperthermia are discussed
together in this section.
CCD is closely associated with dominant RYR1
mutations, whereas MmD is genetically more hetero-
geneous. Most cases of MmD have been attributed to
recessive mutations in RYR1
1618
, SEPN1 (also known
as SELENON)
19
or — less frequently — MYH7
20
.
Histopathological features consistent with MmD have
also been described in some patients with recessive
mutations in MEGF10, which encodes multiple epider-
mal growth factor-like domains protein 10
21–24
. Cores or
minicores in muscle biopsy samples can also be promi-
nent in TTN-related myopathies
25
, often in conjunction
with other myopathic and dystrophic features, and might
occur in other neuromuscular disorders.
Clinically, CCD due to dominant RYR1 mutations
12
is usually a mild condition, although early severe pres-
entations, often associated with denovo inheritance, have
been recorded
26
. Extraocular muscles are usually spared,
and facial, bulbar and respiratory involvement is typically
mild. Congenital dislocation of the hips and scoliosis are
common. Most patients achieve independent ambulation
and have a static or only slowly progressivecourse.
Author addresses
1
Department of Paediatric Neurology, Neuromuscular Service, Evelina’s Children
Hospital, Guy’s and St Thomas’ Hospital NHS Foundation Trust, London, UK.
2
Randall Division of Cell and Molecular Biophysics, Muscle Signalling Section, King’s
College, London, UK.
3
Department of Clinical and Basic Neuroscience, Institute of Psychiatry, Psychology
andNeuroscience (IoPPN), King’s College, London, UK.
4
Departments of Anesthesia and Biomedicine, Basel University and Basel University
Hospital, Basel, Switzerland.
5
Department of Life Sciences, Microbiology and Applied Pathology Section, University
ofFerrara, Ferrara, Italy.
6
The Dubowitz Neuromuscular Centre, Developmental Neurosciences Programme,
UCLGreat Ormond Street Institute of Child Health and Great Ormond Street Hospital
for Children, London, UK.
7
Centre of Human and Aerospace Physiological Sciences, Faculty of Life Science and
Medicine, King’s College, London, UK.
8
NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK.
Key points
Congenital myopathies are clinically and genetically heterogeneous conditions
characterized by muscle weakness and distinctive structural abnormalities in muscle
biopsy samples
Clinically, congenital myopathies have a stable or slowly progressive course, and the
severity varies depending on the causative mutation
More than 20 genes have been implicated in congenital myopathies
The most commonly affected genes encode proteins involved in skeletal muscle Ca
2+
homeostasis, excitation–contraction coupling and thin–thick filament assembly and
interactions
Management of congenital myopathies is largely supportive, although experimental
therapeutic approaches are reaching the clinical trial stage
REVIEWS
152
|
MARCH 2018
|
VOLUME 14 www.nature.com/nrneurol
The clinical features of MmD are more variable
12
.
SEPN1-related myopathies
19,27
are characterized by
marked weakness, early spinal rigidity, scoliosis and res-
piratory impairment. Patients with recessively inherited
RYR1-related core myopathies exhibit a distribution of
weakness and wasting that resembles the SEPN1-related
form but have additional extraocular muscle involve-
ment and, with few exceptions, lack severe respiratory
impairment
17,18
. Various combinations of scoliosis, spinal
rigidity, multiple (mainly distal) contractures and associ-
ated cardiomyopathy can occur in TTN-related and
MYH7‑related forms
20,25
. MEGF10-related myopathies
have a wide clinical spectrum, ranging from a severe
early-onset myopathy with areflexia, respiratory dis-
tress and dysphagia (termed EMARDD)
21,23,24
toadult-
onset cases with minicores in muscle biopsy samples
22
.
Muscle MRI can help to differentiate genetically distinct
coremyopathies
28,29
.
Dominant RYR1-related CCD is allelic to the malig-
nant hyperthermia susceptibility (MHS) trait — a
pharmaco genetic predisposition to malignant hyperther-
mia and severe adverse reactions to volatile anaesthetics
and muscle relaxants
30
— and some CCD-associated
RYR1 mutations also carry an increased risk of MHS. The
association between MHS and recessive RYR1-related
MmD is less well established; however, some cases
of MmD have been attributed to compound hetero-
zygosity for dominant MHS-associated RYR1 muta-
tions and missense, nonsense or other loss-of-function
mutations
18,31,32
.
RYR1-related King–Denborough syndrome (KDS)
is an MHS-associated myopathy characterized by dys-
morphic facial features, short stature, spinal rigid-
ity, scoli osis and various histopathological features
33
.
Another recently recognized myopathy with similar
clinico pathological features is Native American myo-
pathy (NAM), originally described in the Lumbee popu-
lation of North Dakota and caused by homozygosity for a
founder mutation (Trp284Ser) in STAC3, which encodes
SH3 and cysteine-rich domain-containing protein3
34
.
MHS-associated RYR1 mutations have also been
identified as a common cause of exertional myalgia and
rhabdomyolysis (ERM) in otherwise healthy individu-
als with various muscle biopsy findings
35
. Of note, exer-
tional myalgia can be prominent in CCD
36
, and mild to
moderate creatine kinase elevations (up to 1,000 inter-
national units (IU)/l), which are unusual in the context
of other congenital myopathies, are not uncommon.
MHS-associated RYR1 mutations can also give rise to
late-onset axial myopathy in previously healthy (or even
athletic) individuals
37,38
.
Centronuclear myopathy
CNM
39
is associated with X-linked recessive mutations
in the myotubularin gene MTM1 (a condition termed
X-linked myotubular myopathy or XLMTM)
40
, auto-
somal dominant mutations in dynamin 2 (DNM2)
41
and amphiphysin II (BIN1)
42
, and autosomal recessive
mutations in RYR1
43
, BIN1
44
and TTN
45
. Recessive muta-
tions in SPEG have been identified in a small number of
families
46
, and dominant mutations in CCDC78
47
were
found in one isolated pedigree. Heterozygous missense
variants in MTMR14, which have been identified in two
patients with CNM, might represent a genetic modifier
of other genetic backgrounds
48
.
In MTM1-related CNM, the central nuclei are usu-
ally spaced out along the long fibre axis, whereas in
DNM2-related cases, these nuclei can form chains.
Inthe rare BIN1-related cases, the central nuclei can
form clusters. In patients with MTM1-related CNM,
typical features of the muscle fibres include central
areas of increased oxidative enzyme activity and a pale
peripheral halo. These features, along with the presence
of central nuclei, are also seen in congenital myotonic
dystrophy. Strictly centralized nuclei are more common
than multiple internalized nuclei in the MTM1‑related,
DNM2-related and BIN1-related forms of CNM
40,41,44
,
whereas multiple internalized nuclei are more common
in RYR1-related and TTN-related cases
43
. A radial distri-
bution of sarcoplasmic strands that stain positively with
NADH tetrazolium reductase and periodic acid–Schiff
is often seen in DNM2-related CNM
41
. ‘Necklace’ fibres
are often seen in patients carrying milder MTM1 muta-
tions or in female carriers of MTM1 mutations
49
,and
occasionally in patients carrying DNM2 mutations
50
.
Ultrastructural triad abnormalities are observed in most
forms ofCNM
51
.
From a clinical perspective, extraocular muscle
involvement is a consistent feature of most forms of
CNM
52
, the exceptions being the TTN-related, SPEG-
related and CCDC78-related forms. The most severe
form, XLMTM, typically manifests in affected males
with profound hypotonia, weakness and contractures
at birth, as well as bulbar and respiratory involvement
that almost always necessitates ventilation for survival.
Although the provision of constant respiratory support
improves life expectancy in patients with XLMTM,
some long-term survivors experience complica-
tions
53
, probably related to the ubiquitous role of the
defectiveprotein.
Dominantly inherited CNM associated with muta-
tions in DNM2 is frequently a relatively mild condi-
tion
41,54
, although more-severe denovo cases have been
recorded
55,56
. Additional characteristic features of this
condition include distal weakness, calf muscle hyper-
trophy, exertional myalgia and/or fatigue, PNS or CNS
involvement, and multisystem features such as neutro-
penia or cataracts. The peripheral axonal neuropathy
Charcot–Marie–Tooth disease, dominant intermediateB
(CMTDIB) is allelic to DNM2-relatedCNM
57
.
Recessively inherited and — less frequently —
dominantly inherited as well as milder forms of
BIN1-related CNM have been reported in a few fam-
ilies
42,44,58
. Recessively inherited CNM due to RYR1
mutations
43
shows considerable clinical overlap with
other forms of recessively inherited RYR1-related myo-
pathy (see above). Mutations in TTN are often associ-
ated with dysmorphic facial features, scoliosis, spinal
rigidity and contractures
45
, showing some overlap with
Emery–Dreifuss muscular dystrophy and the KDS spec-
trum. Cardiac involvement has been reported only in the
TTN-related and SPEG-related forms ofCNM.
REVIEWS
NATURE REVIEWS
|
NEUROLOGY VOLUME 14
|
MARCH 2018
|
153
Nemaline myopathy
Nemaline myopathy has been associated with muta-
tions in more than ten genes to date, most commonly,
recessive mutations in NEB
59,60
and — usually denovo
— dominant mutations in ACTA1
61
. Rarer causes of
nemaline myopathy, some of which are limited to single
families, include dominant mutations in tropomyosin3
(TPM3)
62
, tropomyosin 2 (TPM2)
63
and KBTBD13
64
and recessive mutations in ACTA1
65
, TPM3
66
, TPM2
67
,
TNNT1
68
, CFL2
69
, KBTBD13
64
, KLHL40
70
, KLHL41
71
,
LMOD3
72
, MYPN
73,74
and MYO18B
75
.
The number and distribution of nemaline rods
vary among muscles and patients. Rods are believed
to be derived from Z-lines and may show continuity
with these structures. The rods are mainly cytoplasmic
but can also be nuclear, particularly in ACTA1-related
Nature Reviews | Neurology
a b
c
d
e
f
g
h
i
j
k
l
m
n
o
100 µm
100 µm
100 µm
100 µm
100 µm
100 µm
10 µm
100 µm
100 µm
100 µm
100 µm
10 µm
10 µm
100 µm
100 µm
REVIEWS
154
|
MARCH 2018
|
VOLUME 14 www.nature.com/nrneurol
nemaline myopathy
76
, where additional actin accumu-
lation and compensatory expression of cardiac actin
can be observed. Nemaline rods are usually seen in
both typeI and typeII muscle fibres, except in patients
with TPM3 mutations, where they are limited to typeI
fibres. Numerous small rectangular rods in fibres with
sparse myofibrils are a feature of KLHL40-related
nemalinemyopathy
70
.
Clinically, nemaline myopathy is highly variable and
is conventionally classified by age of onset and sever-
ity. Severe, often lethal cases within the fetal akinesia
spectrum have been reported in association with reces-
sive mutations in KLHL40
70
, KLHL41
71
, LMOD3
72
and
MYO18B
75
, whereas the typical congenital form charac-
terized by infantile onset, hypotonia and often dispro-
portionate bulbar involvement is most commonly due
to recessive NEB mutations
77
. Dominant — frequently
denovo ACTA1 mutations are often associated with
severe congenital presentations, but milder cases have
been reported
65,78–80
. KBTBD13-related nemaline myo-
pathy is an unusual form characterized by progressive
proximal and neck weakness, gait abnormalities, poor
exercise tolerance and peculiar slowness of movement
81
.
Extraocular muscle involvement is seen in only a frac-
tion of patients with KLHL40, KLHL41 and LMOD3
mutations. Cardiomyopathy is sometimes seen in
MYPN-associated and MYO18B-associated nemaline
myopathy
74,75
. Marked distal involvement is observed
in numerous forms of nemaline myopathy, and many
of the causative genes have also been implicated in dis-
tinct distal arthrogryposis syndromes
82
. Muscle MRI
might help to distinguish different genetic forms of
nemalinemyopathy
83
.
Other congenital myopathies
In recent years, we have witnessed an expansion of
the phenotypic spectrum associated with the known
congenital myopathy-associated genes, as well as the
description of novel conditions that share some of
the clinical and muscle biopsy findings of the better-
characterized entities without reaching a comparable
level of histopathological ‘purity’. These congenital
myopathies with nonspecific, multiple (structural) and
unusual or other features are summarized in the
sections thatfollow.
Congenital myopathies with nonspecific features.
Marked typeI fibre predominance or uniformity is com-
mon in all congenital myopathies and can be the sole
presenting feature
84
. TypeI predominance and atrophy
were also reported in one consanguineous family with
clinical features of a congenital myopathy and reces-
sive mutations in 3-hydroxyacyl-CoA dehydratase1
(HACD1)
85
. Recessive mutations in the corresponding
canine gene cause a form of CNM in dogs
86,87
, although
increased numbers of central nuclei are not a feature in
humans with HACD1 mutations. Congenital fibre type
disproportion, in which typeI fibres are substantially
smaller than typeII fibres, is another common feature
that has been reported in association with mutations in
TPM3
88,89
, RYR1
90
, ACTA1
91
, SEPN1
92
and MYH7
93
, with
or without additional structural abnormalities.
Congenital myopathies with multiple structural abnor-
malities. Congenital myopathies with multiple structural
abnormalities, which were already recognized in the
pre-molecular era
94
, have now been largely genetically
resolved and are often attributed to previously identi fied
genetic backgrounds. The common occurrence of cores
and rods (core–rod myopathy) has been ascribed to
mutations in RYR1, ACTA1 and NEB, whereas the com-
bination of cores and central nuclei is seen with RYR1,
TTN, CCDC78, DNM2 and SPEG mutations.
Novel entities that lack a single predominant histo-
pathological abnormality and, therefore, do not readily
fit into the conventional classification are increasingly
recognized. CACNA1S-related myopathy
95
is character-
ized by marked neonatal hypotonia, generalized weak-
ness with pronounced axial involvement, and variable
extraocular, bulbar and respiratory features. This con-
dition is caused by recessive and dominant mutations
in CACNA1S, which encodes voltage-dependent L-type
Ca
2+
channel subunit-α1S (Cav1.1), the pore-form-
ing subunit of the voltage sensing L-type Ca
2+
channel
dihydro pyridine receptor (DHPR) in skeletal muscle.
Allelic DHPR mutations were previously associated
with dominantly inherited forms of periodic paralysis
(and, in rare cases, MHS phenotypes)
96,97
. Characteristic
histopathological features of CACNA1S-related myo-
pathy include sarcoplasmic reticulum (SR) dilatation,
increased numbers of internal nuclei, and myofibrillar
disorganization resembling minicores.
Recessively inherited PYROXD1-related congenital
myopathy
98
is an early-onset myopathy of moderate
severity characterized by slowly progressive generalized
Fig. 1
|
Muscle pathology in congenital myopathies. Tissue samples from a child with
dominant RYR1-related central core disease (parts ac). Muscle shows myopathic fibre
size variation and marked perimysial fatty infiltration (part a). Most fibres contain a single
central or eccentric core with a well-delineated zone of diminished or absent oxidative
staining; some fibres also show a rim of increased oxidative staining surrounding the core
lesion (part b). Fibres are uniformly typeI (part c). Tissue samples from an adolescent with
recessive SEPN1-related multi-minicore disease (parts df). Muscle shows myopathic fibre
size variation and perimysial fatty infiltration (part d). Fibre typing is preserved, with a
predominance of typeI fibres (darker staining), and both typeI and typeII fibres display
foci of diminished or absent oxidative staining (multi-minicores) and, occasionally, larger
lesions (parts e,f). Tissues samples from a patient with MTM1‑related centronuclear
myopathy (CNM) (parts gi). Samples from a male neonate with severe X-linked recessive
myotubular myopathy show many fibres with centrally placed nuclei (part g). Most fibres
display pale peripheral haloes (part h), and typeI fibres predominate (part i). Tissues
samples from an adult with DNM2-related CNM (parts jl). Muscle shows a marked
increase in central nucleation and perimysial fatty infiltration (part j). Many fibres display
‘radial strands’ emanating from a centrally placed nucleus (part k). TypeI fibre
predominance and hypotrophy create fibre size disproportion; central nuclei are present
in both fibre types (part l). Tissue samples from a severely affected neonate with denovo
dominant ACTA1-related nemaline myopathy (parts mo). Muscle shows myopathic fibre
size variation with the appearance of two fibre populations: smaller typeI and larger
typeII fibres (part m). Numerous thread-like inclusions are seen in both fibre sizes and
appear red with the modified Gomori trichrome stain (part n) and eosinophilic with
haematoxylin and eosin (part m). Pale-stained typeI fibres are often more severely
affected than typeII fibres, showing atrophy or hypotrophy (part o). Muscle biopsy
samples were stained with haematoxylin and eosin (parts a,d,g,j,m), NADH tetrazolium
reductase (parts b,e,h,k) and modified Gomori trichrome (part n), as well as stains for slow
myosin heavy chain (parts c,f,i,l) and myosin ATPase at pH 4.6 (part o).
REVIEWS
NATURE REVIEWS
|
NEUROLOGY VOLUME 14
|
MARCH 2018
|
155
Table 1
|
Genes implicated in congenital myopathies and related conditions
Gene
symbol
Chromosomal
location
Protein Condition Inheritance
Sarcoplasmic reticulum Ca
2+
release, excitation–contraction coupling and/or triadic assembly
RYR1
a
19q13.1 Ryanodine receptor 1 (skeletal) CCD
a
AD, AR
MmD
a
AD, AR
CNM AR
CFTD AR
KDS AR, AD
STAC3 12q13.3 SH3 and cysteine-rich
domain-containing protein 3
NAM AR
ORAI1 12q24.31 Ca
2+
-release-activated Ca
2+
channel protein 1
TAM AD
STIM1 11p15.4 Stromal interaction molecule 1 TAM AD
Stormorken syndrome AD
MTM1
a
Xq28 Myotubularin XLMTM X-linked
BIN1
a
2q14 Amphiphysin II CNM
a
AR, AD
DNM2
a
19p13.2 Dynamin 2 CNM AD
SPEG 2q35 Striated muscle preferentially
expressed protein kinase
Congenital myopathy with central
AR
CCDC78 16p13.3 Coiled-coil domain-containing
protein 78
Congenital myopathy with cores and
central nuclei
AD
CACNA1S 1q32 Voltage-dependent L-type Ca
2+
channel subunit-α1S
Congenital myopathy with EOM AD, AR
SEPN1
a
1p36.13 Selenoprotein N MmD
a
AR
CFTD AR
Thin–thick filament assembly and/or interaction, myofibrillar force generation and protein turnover
NEB
a
2q22 Nebulin Nemaline myopathy
a
AR
ACTA1
a
1q42.1
Actin, α-skeletal muscle
Nemaline myopathy
a
AD, AR
CFTD AD, AR
Cap myopathy AD, AR
TNNT1 19q13.4 Troponin T, slow skeletal muscle Nemaline myopathy AR
TPM2
a
9p13
Tropomyosin β-chain
Nemaline myopathy
a
AD
Cap myopathy AD
DA1A AD
DA2B AD
Escobar syndrome AR
TPM3
a
1q21.2
Tropomyosin α3-chain
Nemaline myopathy
a
AD
CFTD AD
Cap myopathy AD
MYH2 17p13.1 Myosin 2 Congenital myopathy with EOM AD, AR
MYH3 17p13.1 Myosin 3 DA2A, DA2B and DA8 AD
MYH7 14q12 Myosin 7 CFTD AD
MmD AR
MSM AR
MYH8 17p13.1 Myosin 8 Trismus–pseudocamptodactyly
syndrome
AD
Carney complex AD
KBTBD13 15q22.31 Kelch repeat and BTB
domain-containing protein 13
Nemaline myopathy with cores AD
REVIEWS
156
|
MARCH 2018
|
VOLUME 14 www.nature.com/nrneurol
weakness, facial and bulbar involvement, and increased
internalized nuclei and myofibrillar disorganization in
muscle biopsy samples.
Hereditary myosin myopathies (myosinopathies
99
)
comprise distinct distal arthrogryposis syndromes
caused by dominant mutations in MYH3 and MYH8
(which encode two developmental myosin heavy chain
isoforms), as well as congenital myopathies of variable
onset and severity caused by dominant and recessive
mutations in MYH2 and MYH7. MYH7 mutations are
also implicated in Laing distal myopathy and myosin
storage myopathy. In addition to the variable presence of
cores in muscle biopsy samples, recessive MYH2-related
myopathies
100102
are characterized by marked reduction
or absence of typeIIA fibres
99,103
, whereas accumulation
of slow myosin (so-called ‘hyaline bodies’) can be seen
in some MYH7-related cases. Other features that may be
seen in MYH7-related and MYH2-related myopathies
include increased connective tissue, internal nuclei,
rimmed vacuoles, and ring and lobulated fibres
20,93,99,103
.
In the context of overlapping histopathological fea-
tures, the presence of extraocular muscle involve-
ment might cause diagnostic confusion with recessive
RYR1-relatedMmD.
Two other conditions combining ocular involve-
ment, contractures within the distal arthrogryposis
spectrum and features of a congenital myopathy are
recessively inherited ECEL1-related congenital myo-
pathy
104108
and dominantly inherited PIEZO2-related
congenital myopathy
109
(also classified as distal
Table 1 (cont.)
|
Genes implicated in congenital myopathies and related conditions
Gene
symbol
Chromosomal
location
Protein Condition Inheritance
Thin–thick filament assembly and/or interaction, myofibrillar force generation and protein turnover (cont.)
KLHL40
a
2p22.1 Kelch-like protein 40 Nemaline myopathy
a
AR
KLHL41 2q31.1 Kelch-like protein 41 Nemaline myopathy AR
LMOD3 3p14.1 Leiomodin 3 Nemaline myopathy AR
MYBPC3 11p11.2 Myosin binding protein C,
cardiac-type
Congenital myopathy with
cardiomyopathy
AR
MYPN 10q21.3 Myopalladin Nemaline myopathy with
cardiomyopathy
AR
TTN
a
2q31 Titin CNM
a
AR
MmD AR
Other cellular processes or unknown protein functions
CFL2 14q12 Cofilin 2 Nemaline myopathy with cores AR
CNTN1 12q11–q12 Contactin 1 Congenital myopathy lethal AR
ECEL1 2q37.1 Endothelin-converting
enzyme-like 1
DA5 AR
PIEZO2 18p11.21–p22 Piezo-type mechanosensitive
ion channel component 2
Marden–Walker syndrome AD
DA3 AD
DA5 AD
DA with impaired proprioception AR
MEGF10 5q23.2 Multiple epidermal growth
factor-like domains protein 10
Congenital myopathy with minicores AR
Congenital myopathy with areflexia,
respiratory distress and dysphagia
AR
HACD1 10p12.33 Very-long-chain
(3R)-3-hydroxyacyl-CoA
dehydratase 1
Congenital myopathy (nonspecific) AR
SCN4A 17q23.3 Sodium channel, protein type 4
subunit-α
Congenital myopathy (nonspecific) AR
TRIM32 9q33.2 E3 ubiquitin–protein ligase
TRIM32
Sarcotubular myopathy AR
PYROXD1 12p12.1 Pyridine nucleotide-
disulfide oxidoreductase
domain-containing protein 1
Congenital myopathy (nonspecific) AR
AD, autosomal dominant; AR, autosomal recessive; CCD, central core disease; CFTD, congenital fibre type disproportion; CNM,
centronuclear myopathy; DA, distal arthrogryposis; EOM, extraocular muscle involvement; KDS, King–Denborough syndrome;
MmD, multi-minicore disease; MSM, myosin storage myopathy; NAM, North American myopathy; TAM, tubular aggregate
myopathy; XLMTM, X-linked myotubular myopathy.
a
Genes most commonly implicated in the classic structural congenital
myopathies, and the corresponding conditions.
REVIEWS
NATURE REVIEWS
|
NEUROLOGY VOLUME 14
|
MARCH 2018
|
157
arthrogryposis type 5), both of which are associated
with cores and increased internal nuclei in muscle
biopsy samples.
A recessive congenital myopathy due to homozygous
or compound heterozygous mutations in SCN4A, a gene
previously associated with dominantly inherited myo-
tonia and periodic paralysis, was recently described
110
.
This condition has a wide clinical spectrum, from
severe inutero (often early lethal) presentations to
neonatal- onset conditions of variable severity. The
pheno type is mainly characterized by hypotonia, facial
and neck weakness, respiratory and swallowing difficul-
ties and early-onset spinal deformities, but interestingly,
it is not associated with clinical or electrophysiological
evidence of myotonia. Mutations in the same gene are
associated with a presentation featuring severe neo natal
laryngospasm
111
. Histopathologically, SCN4A-related
congenital myopathy is characterized by a combina-
tion of increased fibre size heterogeneity and variable
increases in fatty tissue and tends to lack more-distinct
structural abnormalities
110
. In fact, many of the genetic
backgrounds implicated in congenital myopathies — in
particular, mutations in RYR1, TTN and DNM2 are
associated with marked increases in fat and connective
tissue, mimicking a congenital muscular dystrophy
112,113
.
Congenital myopathies with unusual or other features.
Some of the genes that are associated with nemaline
myopathy — namely, TPM2, TPM3, ACTA1, NEB and
MYPN — have also been implicated in rare myopathies
with unusual histopathological features, such as cap
myopathy and zebra body myopathy
73,114116
.
STIM1-related and ORAI1-related congenital myo-
pathies
117
caused by dominant gain-of-function mutations
result in either tubular aggregate myopathy — a slowly
progressive myopathy with varying degrees of extra-
ocular muscle involvement, exertional myalgia and vari-
able calf hypertrophy — or York platelet and Stormorken
syndromes, related disorders that form a clinical con-
tinuum and are characterized by congenital myopathy,
pupillaryand platelet abnormalities and vari able multi-
system involvement. Recessive inheritance of loss-of-
function mutations in ORAI1 and STIM1 leads to various
combinations of severe combined immunodeficiency,
Table 2
|
Features associated with different genetic backgrounds in congenital myopathies
Feature RYR1
autosomal
dominant
RYR1
autosomal
recessive
SEPN1 TTN MTM1 DNM2 NEB ACTA1 KLHL40
Epidemiology
Frequency of mutations +++ +++ ++ ++ ++ + ++ ++ +
Onset
Infancy ++ +++ + +++ +++ + +++ ++ +++
Childhood +++ ++ +++ + + + + ++ +
Adulthood ++ + +++
Clinical features
Extraocular muscle
involvement
+ +++ +++ +++ ++
Bulbar involvement + +++ ++ ++ +++ ++ ++ ++ +++
Distal involvement + ++ + +++ ++ + +
Respiratory involvement + ++ +++ ++ +++ + ++ ++ +++
Cardiac involvement + +
a
+++
b
+
Contractures + + + +++ +++ ++ ++ ++ +++
Histopathology
Cores +++ +++ +++ ++ + + +
Central nuclei ++ ++ +++ +++ +++
Nemaline rods + + + +++ +++ +++
Fibre type disproportion + +++ + + + +
Connective tissue and/
or fat infiltration
++ ++ ++ +++ +
Imaging
Muscle MRI (specificity
for genetic defect)
+++ ++ ++ + +++ +++ +
Key: –, not reported; +, infrequent; ++, common; +++, very common.
a
Right ventricular impairment secondary to respiratory
involvement.
b
Includes both congenital cardiac defects and acquired cardiomyopathies.
REVIEWS
158
|
MARCH 2018
|
VOLUME 14 www.nature.com/nrneurol
ectodermal dysplasia and congenital myopathy, a com-
bination that was reported in the pre-molecular era in
association with minicores in muscle biopsy samples
118
.
‘Triadin knockout syndrome, which is caused by
compound heterozygosity for TRDN null mutations, is
a recessive cardiac arrhythmia syndrome with various
clinical and histopathological features of a congenital
myopathy, the latter features being characterized by
focal dilatation and degeneration of the lateral SR cis-
ternae
119,120
. The reason for the highly variable pene-
trance of the myopathy associated with TRDN mutations
remains unknown.
Mutations in TRIM32 are associated with limb-girdle
muscular dystrophy type2H and sarcotubular myopathy,
and mutations in TRIM54 and TRIM63 are associated
with microtubular abnormalities and myosin-containing
inclusions
121
. These observations illustrate the increas-
ingly fluid boundaries between congenital myopathies
and other neuromuscular disorders, in particular,
myofibrillar, protein aggregate and vacuolar myopathies.
Pathogenesis
The vast majority of the proteins implicated in congenital
myopathies have been associated with primary or sec-
ondary defects of muscle ECC, intracellular Ca
2+
homeo-
stasis and disturbed sarcomeric assembly and function
(FIG.2), although other mechanisms are emerging.
ECC, muscle contraction and relaxation
ECC is the process whereby an electrical signal gener-
ated by a neuronal action potential is converted into a
chemical gradient — that is, an increase in myoplasmic
Ca
2+
— leading to muscle contraction. The two main
players in skeletal muscle ECC are RYR1 and DHPR
(FIG.2). RYR1 is located on the SR junctional face mem-
brane, and DHPR is located on the plasmalemma and
transverse tubules (T-tubules) — plasmalemmal invagi-
nations that run deep into the muscle fibre. ECC is
extremely rapid, occurring within a few milliseconds,
and relies on a highly defined subcellular architecture,
with each DHPR positioned opposite an RYR1, and
every other RYR1 tetramer facing four DHPRs arranged
in a characteristic checkerboard shape called atetrad.
In addition to its principal regulation through direct
interaction with DHPR, RYR1 is regulated by Ca
2+
and
Mg
2+
and is subjected to post-translational modifications
such as phosphorylation, sumoylation and nitrosylation,
which affect the channel open probability. The junc-
tional SR membrane contains RYR1 as well as many
other smaller proteins, including the structural proteins
triadin and junctin (also known as aspartyl/ asparaginyl
β-hydroxylase), junctional SR protein 1 (JP45), the
high-capacity, low-affinity Ca
2+
binding protein calse-
questrin
122,123
(in an area adjacent to RYR1), and other
proteins with roles in the fine regulation of SR Ca
2+
release or in maintaining the structural integrity of the
Ca
2+
release machinery
122,124131
.
Following its release from the SR, Ca
2+
binds to
troponin C and interacts directly with thin filaments.
As a consequence, muscle contraction occurs in the
sarcomere, a structure that is principally composed of
parallel thick and thin filaments. Sarcomeric regulation
of contraction involves structural changes in the thin
filament complex (composed of actin, tropomyosin and
troponin), triggered by Ca
2+
binding to troponin. The
simplest model for the regulation of the sarcomere by
Ca
2+
is based on steric blocking, whereby tropomyosin
prevents myosin from binding to the actin filament to
generate force. Binding of Ca
2+
to troponin triggers a
chain reaction that results in azimuthal movements of
tropomyosin around the filament to unmask binding
sites on actin for myosin — the molecular motor and
also the main component of thick filaments — allowing
force production and motion
132
. These contractile pro-
teins and related isoforms are differently expressed in
slow-twitch and fast-twitch muscles to fulfil different
functional demands
132
.
Termination of the contraction cycle and muscle
relaxation is achieved by RYR1 closure and activation
of sarcoplasmic/endoplasmic reticulum Ca
2+
ATPase
(SERCA), the protein component that is responsible for
pumping the Ca
2+
back into the SR
133
. SERCA activity is
modulated by two small regulatory proteins, sarcolipin
and phospholamaban
134136
.
Although skeletal muscle ECC can occur in the pres-
ence of extracellular Ca
2+
in the nanomolar range, a wide
consensus exists that Ca
2+
entry from the extracellular
space is essential to ensure prolonged muscle activity.
Two main mechanisms of Ca
2+
entry have been identi-
fied in skeletal muscle: excitation-coupled Ca
2+
entry via
DHPR, which is activated by a train of action potentials or
prolonged membrane depolarization; and store-operated
Ca
2+
entry via stromal interaction molecule 1 (encoded
by STIM1) and Ca
2+
-release-activated Ca
2+
channel
protein 1 (ORAI1), which is triggered by depletion of
endoplasmic reticulum and SR Ca
2+
stores
137–141
.
ECC and Ca
2+
homeostasis abnormalities
Mutations in RYR1 are the most common cause of pri-
mary defects of ECC and Ca
2+
homeostasis
7,18,142,143
.
Functional studies utilizing cellular and animal
models
144,145
indicate that excessive Ca
2+
release and
lower RYR1 activation thresholds are consequences
of dominantly inherited MHS-associated RYR1 muta-
tions, whereas both SR Ca
2+
store depletion with a
resulting increase in cytosolic Ca
2+
levels (the ‘leaky
channel’ hypothesis) and disturbed ECC (the ‘excitation–
contraction uncoupling’ hypothesis) have been proposed
as mechanisms underlying dominantly inherited CCD
142
.
On the basis of the limited studies performed so far,
quantitative reduction of RYR1 channels is a more likely
mechanism than qualitative RYR1 dysfunction in reces-
sive RYR1-related myopathies
146148
reductions in Cav1.1 protein levels are seen in recessive
RYR1-related and CACNA1S-related congenital myo-
pathies
95,146
; the latter conditions also feature disturbed
ECC and, consequently, reduced depolarization- induced
SR Ca
2+
release in myotubes and mature muscle fibres.
STAC3, the gene that is homozygously mutated in NAM,
encodes a protein that targets Cav1.1 to the T-tubules and,
thus, participates in voltage-induced Ca
2+
release
149,150
.
Disturbances in voltage-induced Ca
2+
release are likely
REVIEWS
NATURE REVIEWS
|
NEUROLOGY VOLUME 14
|
MARCH 2018
|
159
to be involved in the recently described triadin knockout
syndrome
119
, although the basis for the highly variable
penetrance of skeletal muscle features in this condition
is currentlyuncertain.
Dominant mutations in STIM1 and ORAI1 are
associated with distinct alterations in store-operated
Ca
2+
influx, resulting in increased resting Ca
2+
levels
due to mediation of Ca
2+
influx by constitutively active
molecules independently of SR Ca
2+
levels
140,151
. By con-
trast, recessive ORAI1 mutations, which lead to reduced
ORAI1 expression, result in impaired Ca
2+
influx
152
.
Secondary defects of ECC and Ca
2+
homeostasis,
probably due to RYR1 redox modifications, have been
demonstrated in SEPN1-mutated myotubes and in the
Sepn1-knockout mouse model
153,154
. Many of the genes
implicated in CNM, including MTM1
40
, DNM2
41
and
Dihydropyridine
receptor
RYR1
SERCA
Phospholamban
Myoregulin
Calsequestrin
Sarcalumenin
JP45
α-Actinin
Myomesin
Actin
Actin filament
Myosin
Titin
Titin
Transverse tubule
SR terminal cisternae Longitudinal SRJFM
Nebulin
Ca
2+
Myosin
Triadin
Z I A M
Junctin
STAC3
Nature Reviews | Neurology
REVIEWS
160
|
MARCH 2018
|
VOLUME 14 www.nature.com/nrneurol
BIN1
44
, encode proteins that have an important role in
intricately linked intracellular membrane trafficking
pathways. Mutations in these genes might indirectly
affect muscle Ca
2+
handling and ECC, probably sec-
ondary to abnormalities of triad assembly and the ECC
machinery
155
. Although such abnormalities have been
demonstrated in mouse models of both DNM2-related
and MTM1-related myopathies
156
, a recent study on
MTM1-mutated human myoblasts failed to demonstrate
any alterations in ECC and Ca
2+
release, indicating that
these alterations may reflect long-term effects invivo
157
.
The pathogenicity of TTN mutations is probably
multifactorial and is likely to involve several mechan-
isms implicated in ECC, including calpain3- mediated
RYR1 recruitment to the triad and obscurin-
mediated interactions between the T-tubules, the SR
and the sarcomere.
Sarcomeric abnormalities
The majority of the genes implicated in nemaline myo-
pathy to date, including NEB
59
, ACTA1
61
, TPM2
63
,
TPM3
62
and TNNT1
68
, are involved in thin filament
assembly and interactions. Pathogenic mutations
in the two most commonly mutated genes, NEB
andACTA1, have been extensively studied
158
. Dominant
ACTA1mutations exert a dominant-negative effect on
muscle function that is mediated through lowered Ca
2+
sensitivity, whereas recessive ACTA1 mutations abol-
ish functional protein expression, with the phenotype
severity probably depending on the expression of com-
pensatory proteins such as actin, α-cardiac muscle1
(ACTC1)
159,160
. In rare cases, ACTA1 mutations result in
increased muscle contractility
161,162
.
NEB mutations affect the specific function of nebu-
lin in thin filament regulation and force generation
163
.
The effects of various nemaline myopathy-associated
mutations on the interactions of nebulin with actin
and tropomyosin, thin filament length and force gen-
eration were demonstrated in two invitro studies
164,165
.
MYO18B, which was found to be mutated in one family
with a severe form of nemaline myopathy
75
, encodes an
unconventional myosin protein with a more general role
in sarcomeric assembly and maintenance
166,167
.
KBTBD13, KLHL40, KLHL41 and LMOD3, which
have recently been implicated in nemaline myopathy,
encode a group of Kelch and Kelch-like proteins that are
not primary thin filament components but are involved
in muscle quality control processes
168
and may, thus,
affect myofibrillar assembly and function indirectly.
Evidence for a direct interaction between Kelch-like
protein 40 (KLHL40), nebulin and leiomodin 3 has
beenprovided
169
.
The myosinopathies
99
— disorders of the thick fila-
ments — are likely to cause muscle disease through two
principal mechanisms: disturbed thick filament inter-
action and function and, particularly in MYH7-related
congenital myopathies
99
, aggregation of abnormal
protein.
Other pathogenic mechanisms
Some of the proteins implicated in congenital myo-
pathies are specifically involved in ECC and Ca
2+
homeo stasis, whereas others are thought to have addi-
tional roles in and beyond muscle. Selenoprotein N
(encoded by SEPN1) belongs to a family of proteins
that contain selenium in the form of the 21st amino
acid, selenocysteine. In muscle, this protein has been
specifically implicated in myogenesis — a role that it
shares with the protein encoded by MEGF10, which is
mutated in a rare form of MmD
23
— and redox regula-
tion
170,171
. The important role of normally functioning
redox regulation for muscle health is also illustrated
by the identification of recessive mutations in the
oxidoreductase- encoding gene PYROXD1 as a cause of
early-onset congenital myopathies
98
.
Reflecting their essential roles in intricately linked
intracellular membrane trafficking pathways, mutations
in the CNM-associated genes MTM1, DNM2 and BIN1
have been associated with a wide range of downstream
effects, including defects in mitochondria, the inter-
mediate filament protein desmin, satellite cell activation
and the neuromuscular junction
155
. Abnormalities of
muscle membrane systems have also been described in
association with canine HACD1-related CNM
86,87
, a natu-
rally occurring animal model of a nonspecific congenital
myopathy that has also been described in humans
172
.
The CNM-associated genes MTM1 and DNM2 have
also been implicated in pathways that affect muscle
protein turnover and/or muscle growth and atrophy
pathways. In zebrafish and mouse models of myotubu-
larin deficiency, disturbances of the autophagy pathway,
associated with F-box only protein 32 upregulation and
atrophy, have been reported
173175
. Abnormalities of
autophagosome maturation and autophagic flux have
also been described in a mouse model of DNM2‑related
CNM in association with marked muscle atrophy
and weakness
176
. Several mechanisms might affect
Fig. 2
|
Proteins involved in congenital myopathies. The figure shows the subcellular
localization of the main proteins implicated in skeletal muscle excitation–contraction
coupling (ECC) and thin–thick filament interaction and assembly. Genes encoding
components of the ECC machinery and the thin and thick filaments of skeletal muscle
are commonly mutated in congenital myopathies. The transverse tubules are
invaginations of the plasma membrane where the dihydropyridine receptor complex
(containing SH3 and cysteine-rich domain-containing protein 3 (STAC3)) is located.
Thismembrane compartment faces the sarcoplasmic reticulum (SR) junctional face
membrane (JFM), which contains ryanodine receptor 1 (RYR1) as well as junctional SR
protein 1 (JP45) and the structural proteins triadin and aspartyl/asparaginyl
β-hydroxylase (junctin). Calsequestrin bound to Ca
2+
forms a mesh-like structure within
the lumen of the SR terminal cisternae. JP45 also interacts with calsequestrin via its
lumenal carboxy-terminal domain. Ca
2+
release into the cytosol results in sarcomeric
shortening through specific interactions between thin and thick filaments, in particular,
the sliding of actin past myosin filaments. ECC is terminated through SR Ca
2+
reuptake
through sarcoplasmic/endoplasmic reticulum Ca
2+
ATPase (SERCA) Ca
2+
pumps. SERCAs
are present in the terminal cisternae as well as the longitudinal SR and are regulated by
phospholamban, myoregulin and sarcolipin. The Ca
2+
-buffering protein sarcalumenin is
also located in the longitudinal SR and terminal cisternae and is involved in regulating
SERCA activity. A, A-band; I, I-band; M, M-band; Z, Z-line. Image courtesy of Christoph
Bachmann, Departments of Anesthesia and Biomedicine, Basel University Hospital,
Basel, Switzerland. 3D representations from RSCB PDB: calsequestrin, PDB ID 2VAF
(REF.227); dihydropyridine receptor, PDB ID 4MS2 (REF. 228); phospholamban, PDB ID
1N7L (REF. 229); RYR1, PDB ID 4UWA (REF. 230); SERCA, PDB ID 1SU4 (REF. 231); STAC3,
PDB ID 2DB6 (http://www.rcsb.org/pdb/explore.do?structureId=2db6).
REVIEWS
NATURE REVIEWS
|
NEUROLOGY VOLUME 14
|
MARCH 2018
|
161
autophagy and other degradation pathways in TTN-
related CNM. These mechanisms include abrogation
of calpain 3-mediated protein turnover (inthe caseof
C-terminal-truncating TTN mutations), perturba-
tion of the link between titin and the ubiquitin ligase
myospryn
177
, and perturbation of the link between the
titin kinase domain and the autophagy cargo adap-
tors NBR1 (next to BRCA1 gene 1 protein NBR1) and
sequestosome 1 (SQSTM1) by M-band-disrupting TTN
mutations
25
. Intriguingly, the typical histopathological
features of CNM have also been reported in primary
disorders of autophagy
178,179
, further supporting a
close link between defective autophagy and abnormal
nuclearpositioning.
A novel epigenetic mechanism involving altera-
tions of muscle-specific microRNAs, increased DNA
methy lation and increased expression of classII histone
deacetylases has been reported in RYR1-related myo-
pathies
180
and might also be relevant for other congenital
myopathies
157
.
The mechanisms through which mutations in
ECEL1, PIEZO2 and SCN4A cause specific early-onset
congenital myopathies are currently uncertain.
Diagnostic approach
The International Standard of Care Committee for
Congenital Myopathies has provided a structured diag-
nostic approach to the congenital myopathies
181
. Many
features, including axially pronounced weakness and
hypotonia, are consistent but nonspecific on clinical
assessment, whereas others — in particular, the degree
of distal, extraocular muscle, cardiac and respiratory
involvement — can indicate specific genetic backgrounds.
Useful laboratory investigations include measure-
ment of serum creatine kinase levels, which are typically
normal or slightly elevated, and assays for acetylcholine
receptor antibodies to exclude autoimmune myasthenic
conditions
182
. Neurophysiological studies, such as elec-
tromyography and nerve conduction studies, are useful
mainly for excluding congenital neuropathies, myotonic
disorders
111
and congenital myasthenic syndromes
183
.
Muscle imaging
6
, in particular, muscle ultrasonog-
raphy as a screening test and muscle MRI for a more
detailed assessment, can reveal diagnostic patterns of
selective muscle involvement. Assessment of muscle
biopsy samples with a standard panel of histological,
histochemical and immunohistochemical stains
13
will
confirm the specific congenital myopathy and exclude
distinct conditions with overlapping pathological fea-
tures, such as the congenital muscular dystrophies
184
,
myofibrillar myopathies
185
and autophagic vacuolar
myopathies
186
. Electron microscopy helps to clarify the
pathognomonic structural abnormalities that are seen
with light microscopy.
Concomitant analysis of multiple congenital myo-
pathy-associated genes through NGS is rapidly becom-
ing the preferred diagnostic approach. Functional studies
will be increasingly relevant for pathogenicity assessment
ofvariants in large genes such as TTN, NEB and RYR1, as
variants of uncertain significance are not uncommon in
these genes, even in healthy control populations.
Management and therapy development
Supportive management
Supportive management (outlined in detail elsewhere
187
)
is based on a multidisciplinary approach. Regular physio-
therapy and provision of orthotic support is beneficial
to prevent contracture development and to maintain
mobility. Patients with dysarthria and/or feeding dif-
ficulties will benefit from regular speech and language
therapy. In some cases, bulbar involvement and poor
weight gain may necessitate gastrostomy. Regular mon-
itoring of respiratory function (including sleep studies)
and proactive respiratory management (including timely
noninvasive ventilation and cough assistance techniques)
are essential, particularly in conditions where substan-
tial respiratory involvement — often disproportionate
to the degree of limb-girdle weakness — is recognized.
Regular cardiac monitoring is crucial for patients with
congenital myopathies that are consistently associated
with cardiomyopathy (in particular, the TTN-related and
MYH7-related forms) and also for individuals in whom
the genetic defect is uncertain. Given the often-complex
comorbidities associated with congenital myopathies,
orthopaedic surgery, most notably to treat scoliosis,
should be undertaken at a tertiary neuro muscular centre.
MHS must be anticipated in the anaesthetic management
of patients with RYR1 or STAC3 mutations and in those
with unresolved genetic backgrounds.
Mechanism-based therapies
Mechanism-based therapies for the congenital myo-
pathies that are already available or currently in
development have been reviewed in detail elsewhere
188
.
Genetic therapies. Owing to their enormous size, most of
the genes commonly implicated in congenital myopathies
are not amenable to virus-based gene transfer approaches.
However, delivery of MTM1 via an adeno-associated
virus 8-based vector has been demonstrated to improve
the clinicopathological phenotype in Mtm1-deficient
mice and a canine model of XLMTM
172,189
.
Restoration of the mRNA reading frame is theoret-
ically applicable to various congenital myopathies in
which nonsense mutations are implicated. Exon skip-
ping has been successfully applied invitro to remove a
pseudo-exon from the mRNA of a child with a recessive
RYR1-related myopathy
190
. Considering that carriers
of truncating RYR1 mutations are asymptomatic
190,191
,
selective silencing of the mutant gene could be a fea-
sible therapeutic strategy for dominant RYR1-related
myopathies in the future. Pharmacological suppres-
sion of stop codons
192
by compounds such as ataluren
is a potential approach in congenital myopathies that
involve nonsense mutations, although it is currently
uncertain whether such an approach will increase
normal protein levels sufficiently to restore structural
integrity and function, and the effects on the many
loss-of-function variants in the human genome have
yet to be determined
193
.
Downregulation or upregulation of genes that act
in convergent pathways could be a relevant approach
for various forms of CNM. Studies have demonstrated
REVIEWS
162
|
MARCH 2018
|
VOLUME 14 www.nature.com/nrneurol
that downregulation of dynamin 2
194
or targeting of
classII and III phosphoinositide 3-kinases in muscle
195
can rescue the phenotype in XLMTM animal models,
suggesting that pharmacological modification of
intricately linked pathways is a potential treatment
modality for XLMTM and, possibly, other forms of
CNM. Upregulation of cardiac actin might be a fea-
sible therapeutic approach for patients with ACTA1
nullmutations
196,197
.
Enzyme replacement therapy. Enzyme replacement
therapy is currently relevant only to XLMTM caused by
loss of myotubularin function. In Mtm1-knockout mice,
improvements in contractile function and histopatho-
logical features were observed following short-term
myotubularin enzyme replacement
198
.
Pharmacological therapies. Pharmacological therapies
that are potentially applicable to congenital myopathies
can be grossly divided into three principal approaches:
direct modification of altered protein function (for
example, modification of RYR1 release in RYR1-related
myopathies); enhancement of thin–thick filament inter-
actions (for example, in some nemaline myopathies);
and therapies aimed at nonspecifically ameliorating
downstream effects of the specific gene mutation.
Modification of RYR1 Ca
2+
release by use of the spe-
cific RYR1 antagonist dantrolene
199
is the established
emergency treatment for malignant hyper thermia
and has also been used effectively in a few patients
with RYR1-related ERM
35,200
and CCD
201,202
. Other
compounds with the potential to treat excessive SR
Ca
2+
release and/or increased SR Ca
2+
leakage are the
calstabin-stabilizing 1,4-benzothiazepine derivatives
JTV519 and S107
203,204
and the AMP-activated protein
kinase activator 5-aminoimidazole-4- carboxamide
ribonucleotide (AICAR)
205,206
. However, the safety pro-
files of these compounds in humans and their roles
in RYR1-related myopathies associated with reduced
rather than increased Ca
2+
conductance are currently
uncertain.
Enhancement of filament interactions and promo-
tion of force production
207,208
, either by slowing the rate
of Ca
2+
release from troponin C or by directly targeting
myosin molecules, are potentially valuable approaches
for some nemaline myopathies. However, concerns
remain regarding fibre type specificity and potential
cardiac adverse effects of the molecules that are being
utilized.
Modification of the downstream effects of spe-
cific gene mutations encompasses various approaches.
Inhibition of myostatin, an important negative regula-
tor of muscle fibre size
209
, might be applicable to con-
genital myopathies in which fibre atrophy is prominent.
Following observations of increased oxidative stress and
a favourable response to these compounds in animal
models
154,210,211
, antioxidants such as N-acetylcysteine
are currently being investigated in clinical trials con-
cerning RYR1-related and SEPN1-related myopathies.
In light of the neuromuscular junction and transmis-
sion abnormalities in CNM, RYR1-related MmD and
KLHL40-related nemaline myopathy
212–215
, acetylcho-
linesterase inhibitors have been used with some benefit
in a small number of patients. Two other compounds,
salbutamol for core myopathies
216–218
and — supported
by preclinical data from a relevant animal model
219
-tyrosine in nemaline myopathy
220
, have shown
apparent benefits in open-label pilot studies.
For those disease entities where misfolded proteins
or domains have unequivocal primary roles in the
disease process (for example, titin in autosomal reces-
sive MmD with heart disease), compounds that act
as chemical chaperones show promise. A pharmaco-
chaperone approach, using the small amphipathic com-
pound 4-phenylbutyrate, was shown to alleviate some
of the pathological features in a mouse model of PLEC-
associated epidermolysis bullosa simplex with muscu-
lar dystrophy
221
, although it is uncertain whether the
observed effect was attributable to stabilization of mis-
folded mutant protein or its clearance through induc-
tion of autophagy by the drug
222,223
. A beneficial effect
of 4-phenylbutyrate has also been suggested in a mouse
model of RYR1-related myopathy
224
. The range of chem-
ical chaperones is increasing rapidly
225
, but the half-
maximal inhibitory concentration — a measure of the
ability of a compound to inhibit a specific function — is
often low
226
, and the development of more target-specific
compounds might make this approach more effective
and applicable.
Conclusions and outlook
Widespread clinical implementation of NGS has rapidly
expanded the genetic and clinicopathological spectrum
of the congenital myopathies. In addition to the classic
entities CCD, MmD, CNM and nemaline myopathy,
the congenital myopathies now encompass a wide range
of early-onset, non-dystrophic neuromuscular dis-
orders with various combinations of structural defects.
Congenital myopathies due to mutations in RYR1, the
most common genetic cause, form a continuum with
intermittent induced myopathies, such as malignant
hyperthermia and exertional rhabdomyolysis, in other-
wise healthy individuals. Other forms of congenital myo-
pathy overlap substantially with the distal arthrogryposis
and protein aggregation myopathy spectrum, particularly
in cases where sarcomeric proteins are implicated.
The unravelling of the underlying molecular mech-
anisms has advanced not only our understanding ofthe
congenital myopathies but also our knowledge of nor-
mal muscle physiology and homeostasis. Although
the primary genetic defects and principal pathogenic
mechanisms have largely been elucidated, downstream
effects on muscle growth and atrophy pathways, the
role of genetic and other modifiers, and the molecular
basis of the common histopathological features remain
uncertain.
Specific therapies for congenital myopathies, utiliz-
ing genetic, enzyme replacement and pharmacological
approaches, are currently being developed or are already
reaching the clinical trial stage, emphasizing the need for
comprehensive natural history studies concerning these
clinically variable conditions.
REVIEWS
NATURE REVIEWS
|
NEUROLOGY VOLUME 14
|
MARCH 2018
|
163
1. Magee,K.R. & Shy,G.M. A new congenital non-
progressive myopathy. Brain 79, 610–621 (1956).
2. Engel,A.G., Gomez,M.R. & Groover,R.V. Multicore
disease. A recently recognized congenital myopathy
associated with multifocal degeneration of muscle
fibers. Mayo Clin. Proc. 46, 666–681 (1971).
3. Spiro,A.J., Shy,G.M. & Gonatas,N.K. Myotubular
myopathy. Persistence of fetal muscle in an adolescent
boy. Arch. Neurol. 14, 1–14 (1966).
4. Shy,G.M., Engel,W.K., Somers,J.E. & Wanko,T.
Nemaline myopathy. a new congenital myopathy. Brain
86, 793–810 (1963).
5. Lopez,R.J. etal. An RYR1 mutation associated with
malignant hyperthermia is also associated with
bleeding abnormalities. Sci. Signal. 9, ra68 (2016).
6. Jungbluth,H. Myopathology in times of modern
imaging. Neuropathol. Appl. Neurobiol. 43, 24–43
(2017).
7. Snoeck,M. etal. RYR1-related myopathies:
a wide spectrum of phenotypes throughout life.
Eur.J.Neurol. 22, 1094–1112 (2015).
8. Jungbluth,H. & Voermans,N.C. Congenital
myopathies: not only a paediatric topic. Curr. Opin.
Neurol. 29, 642–650 (2016).
9. Quane,K.A. etal. Mutations in the ryanodine
receptor gene in central core disease and malignant
hyperthermia. Nat. Genet. 5, 51–55 (1993).
10. Fujii,J. etal. Identification of a mutation in porcine
ryanodine receptor associated with malignant
hyperthermia. Science 253, 448–451 (1991).
11. Biancalana,V. & Laporte,J. Diagnostic use of
massively parallel sequencing in neuromuscular
diseases: towards an integrated diagnosis.
J.Neuromuscul. Dis. 2, 193–203 (2015).
12. Jungbluth,H., Sewry,C.A. & Muntoni,F. Core
myopathies. Semin. Pediatr. Neurol. 18, 239–249
(2011).
13. Dubowitz,V., Sewry,C.A. & Oldfors,A. Muscle
Biopsy: a Practical Approach 4th edn (Saunders,
2013).
14. Amburgey,K. etal. Prevalence of congenital
myopathies in a representative pediatric United
States population. Ann. Neurol. 70, 662–665
(2011).
15. Hackman,P., Udd,B., Bonnemann,C.G., Ferreiro,A.
& Titinopathy Database Consortium. 219th ENMC
International Workshop Titinopathies International
database of titin mutations and phenotypes,
Heemskerk, The Netherlands, 29April−1May 2016.
Neuromuscul. Disord. 27, 396–407 (2017).
16. Jungbluth,H. etal. Autosomal recessive inheritance of
RYR1 mutations in a congenital myopathy with cores.
Neurology 59, 284–287 (2002).
17. Jungbluth,H. etal. Minicore myopathy with
ophthalmoplegia caused by mutations in the
ryanodine receptor type1 gene. Neurology 65,
1930–1935 (2005).
18. Klein,A. etal. Clinical and genetic findings in a large
cohort of patients with ryanodine receptor 1 gene-
associated myopathies. Hum. Mutat. 33, 981–988
(2012).
19. Ferreiro,A. etal. Mutations of the selenoprotein N
gene, which is implicated in rigid spine muscular
dystrophy, cause the classical phenotype of
multiminicore disease: reassessing the nosology of
early-onset myopathies. Am. J.Hum. Genet. 71,
739–749 (2002).
20. Cullup,T. etal. Mutations in MYH7 cause multi-
minicore disease (MmD) with variable cardiac
involvement. Neuromuscul. Disord. 22, 1096–1104
(2012).
21. Takayama,K. etal. Japanese multiple epidermal
growth factor 10 (MEGF10) myopathy with novel
mutations: a phenotype–genotype correlation.
Neuromuscul. Disord. 26, 604–609 (2016).
22. Liewluck,T. etal. Adult-onset respiratory insufficiency,
scoliosis, and distal joint hyperlaxity in patients with
multiminicore disease due to novel Megf10 mutations.
Muscle Nerve 53, 984–988 (2016).
23. Logan,C.V. etal. Mutations in MEGF10, a regulator
of satellite cell myogenesis, cause early onset
myopathy, areflexia, respiratory distress and
dysphagia (EMARDD). Nat. Genet. 43, 1189–1192
(2011).
24. Boyden,S.E. etal. Mutations in the satellite cell gene
MEGF10 cause a recessive congenital myopathy with
minicores. Neurogenetics 13, 115–124 (2012).
25. Chauveau,C. etal. Recessive TTN truncating
mutations define novel forms of core myopathy with
heart disease. Hum. Mol. Genet. 23, 980–991
(2014).
26. Romero,N.B. etal. Dominant and recessive central
core disease associated with RYR1 mutations and fetal
akinesia. Brain 126, 2341–2349 (2003).
27. Scoto,M. etal. SEPN1-related myopathies: clinical
course in a large cohort of patients. Neurology 76,
2073–2078 (2011).
28. Klein,A. etal. Muscle MRI in congenital myopathies
due to ryanodine receptor type1 gene mutations.
Arch. Neurol. 68, 1171–1179 (2011).
29. Jungbluth,H. etal. Magnetic resonance imaging of
muscle in congenital myopathies associated with RYR1
mutations. Neuromuscul. Disord. 14, 785–790
(2004).
30. Rosenberg,H., Davis,M., James,D., Pollock,N. &
Stowell,K. Malignant hyperthermia. Orphanet J.Rare
Dis. 2, 21 (2007).
31. Zhou,H. etal. Characterization of recessive RYR1
mutations in core myopathies. Hum. Mol. Genet. 15,
2791–2803 (2006).
32. Kraeva,N. etal. Compound RYR1 heterozygosity
resulting in a complex phenotype of malignant
hyperthermia susceptibility and a core myopathy.
Neuromuscul. Disord. 25, 567–576 (2015).
33. Dowling,J.J. etal. King–Denborough syndrome with
and without mutations in the skeletal muscle
ryanodine receptor (RYR1) gene. Neuromuscul.
Disord. 21, 420–427 (2011).
34. Horstick,E.J. etal. Stac3 is a component of the
excitation–contraction coupling machinery and
mutated in Native American myopathy. Nat. Commun.
4, 1952 (2013).
35. Dlamini,N. etal. Mutations in RYR1 are a common
cause of exertional myalgia and rhabdomyolysis.
Neuromuscul. Disord. 23, 540–548 (2013).
36. Bethlem,J., van Gool,J., Hulsmann,W.C. &
Meijer,A.E. Familial non-progressive myopathy with
muscle cramps after exercise. A new disease
associated with cores in the muscle fibres. Brain 89,
569–588 (1966).
37. Løseth,S. etal. A novel late-onset axial myopathy
associated with mutations in the skeletal muscle
ryanodine receptor (RYR1) gene. J.Neurol. 260,
1504–1510 (2013).
38. Jungbluth,H. etal. Late-onset axial myopathy with
cores due to a novel heterozygous dominant mutation
in the skeletal muscle ryanodine receptor (RYR1) gene.
Neuromuscul. Disord. 19, 344–347 (2009).
39. Jungbluth,H., Wallgren-Pettersson,C. & Laporte,J.
Centronuclear (myotubular) myopathy.
OrphanetJ.Rare Dis. 3, 26 (2008).
40. Laporte,J. etal. A gene mutated in X-linked
myotubular myopathy defines a new putative tyrosine
phosphatase family conserved in yeast. Nat. Genet.
13, 175–182 (1996).
41. Bitoun,M. etal. Mutations in dynamin 2 cause
dominant centronuclear myopathy. Nat. Genet. 37,
1207–1209 (2005).
42. Böhm,J. etal. Adult-onset autosomal dominant
centronuclear myopathy due to BIN1 mutations. Brain
137, 3160–3170 (2014).
43. Wilmshurst,J.M. etal. RYR1 mutations are a
common cause of congenital myopathies with central
nuclei. Ann. Neurol. 68, 717–726 (2010).
44. Nicot,A.S. etal. Mutations in amphiphysin 2 (BIN1)
disrupt interaction with dynamin 2 and cause
autosomal recessive centronuclear myopathy.
Nat.Genet. 39, 1134–1139 (2007).
45. Ceyhan-Birsoy,O. etal. Recessive truncating titin
gene. TTN, mutations presenting as centronuclear
myopathy. Neurology 81, 1205–1214 (2013).
46. Agrawal,P.B. etal. SPEG interacts with myotubularin,
and its deficiency causes centronuclear myopathy with
dilated cardiomyopathy. Am. J.Hum. Genet. 95,
218–226 (2014).
47. Majczenko,K. etal. Dominant mutation of CCDC78 in
a unique congenital myopathy with prominent internal
nuclei and atypical cores. Am. J.Hum. Genet. 91,
365–371 (2012).
48. Tosch,V. etal. A novel PtdIns3P and PtdIns(3,5)P2
phosphatase with an inactivating variant in
centronuclear myopathy. Hum. Mol. Genet. 15,
3098–3106 (2006).
49. Bevilacqua,J.A. etal. “Necklace” fibers, a new
histological marker of late-onset MTM1-related
centronuclear myopathy. Acta Neuropathol. 117 ,
283–291 (2009).
50. Liewluck,T., Lovell,T.L., Bite,A.V. & Engel,A.G.
Sporadic centronuclear myopathy with muscle
pseudohypertrophy, neutropenia, and necklace fibers
due to a DNM2 mutation. Neuromuscul. Disord. 20,
801–804 (2010).
51. Toussaint,A. etal. Defects in amphiphysin 2 (BIN1)
and triads in several forms of centronuclear
myopathies. Acta Neuropathol. 121, 253–266
(2011).
52. Romero,N.B. Centronuclear myopathies: a widening
concept. Neuromuscul. Disord. 20, 223–228 (2010).
53. Herman,G.E., Finegold,M., Zhao,W., de Gouyon,B.
& Metzenberg,A. Medical complications in long-term
survivors with X-linked myotubular myopathy.
J.Pediatr. 134, 206–214 (1999).
54. Bohm,J. etal. Mutation spectrum in the large GTPase
dynamin 2, and genotype–phenotype correlation in
autosomal dominant centronuclear myopathy.
Hum.Mutat. 33, 949–959 (2012).
55. Bitoun,M. etal. Dynamin 2 mutations cause sporadic
centronuclear myopathy with neonatal onset.
Ann.Neurol. 62, 666–670 (2007).
56. Jungbluth,H. etal. Centronuclear myopathy with
cataracts due to a novel dynamin 2 (DNM2) mutation.
Neuromuscul. Disord. 20, 49–52 (2010).
57. Zuchner,S. etal. Mutations in the pleckstrin homology
domain of dynamin 2 cause dominant intermediate
Charcot–Marie–Tooth disease. Nat. Genet. 37,
289–294 (2005).
58. Jungbluth,H., Wallgren-Pettersson,C. & Laporte,J.F.
198th ENMC International Workshop: 7th Workshop
on Centronuclear (Myotubular) Myopathies, 31st
May−2nd June 2013, Naarden, The Netherlands.
Neuromuscul. Disord. 23, 1033–1043 (2013).
59. Pelin,K. etal. Mutations in the nebulin gene
associated with autosomal recessive nemaline
myopathy. Proc. Natl Acad. Sci. USA 96, 2305–2310
(1999).
60. Pelin,K. etal. Nebulin mutations in autosomal
recessive nemaline myopathy: an update.
Neuromuscul. Disord. 12, 680–686 (2002).
61. Nowak,K.J. etal. Mutations in the skeletal muscle
α-actin gene in patients with actin myopathy and
nemaline myopathy. Nat. Genet. 23, 208–212
(1999).
62. Laing,N.G. etal. A mutation in the α tropomyosin
gene TPM3 associated with autosomal dominant
nemaline myopathy. Nat. Genet. 9, 75–79 (1995).
63. Donner,K. etal. Mutations in the β-tropomyosin
(TPM2) gene — a rare cause of nemaline myopathy.
Neuromuscul. Disord. 12, 151–158 (2002).
64. Sambuughin,N. etal. Dominant mutations in
KBTBD13, a member of the BTB/Kelch family, cause
nemaline myopathy with cores. Am. J.Hum. Genet.
87, 842–847 (2010).
65. Laing,N.G. etal. Mutations and polymorphisms of
the skeletal muscle α-actin gene (ACTA1 ). Hum. Mutat.
30, 1267–1277 (2009).
66. Lehtokari,V.L. etal. Identification of a founder
mutation in TPM3 in nemaline myopathy patients of
Turkish origin. Eur. J.Hum. Genet. 16, 1055–1061
(2008).
67. Monnier,N. etal. Absence of β-tropomyosin is a new
cause of Escobar syndrome associated with nemaline
myopathy. Neuromuscul. Disord. 19, 118–123
(2009).
68. Johnston,J.J. etal. A novel nemaline myopathy in
theAmish caused by a mutation in troponin T1.
Am.J.Hum. Genet. 67, 814–821 (2000).
69. Agrawal,P.B. etal. Nemaline myopathy with
minicores caused by mutation of the CFL2 gene
encoding the skeletal muscle actin-binding protein,
cofilin-2. Am. J.Hum. Genet. 80, 162–167 (2007).
70. Ravenscroft,G. etal. Mutations in KLHL40 are a
frequent cause of severe autosomal-recessive
nemaline myopathy. Am. J.Hum. Genet. 93, 6–18
(2013).
71. Gupta,V.A. etal. Identification of KLHL41 mutations
implicates BTB-Kelch-mediated ubiquitination as an
alternate pathway to myofibrillar disruption in
nemaline myopathy. Am. J.Hum. Genet. 93,
1108–1117 (2013).
72. Yuen,M. etal. Leiomodin-3 dysfunction results in thin
filament disorganization and nemaline myopathy.
J.Clin. Invest. 124, 4693–4708 (2014).
73. Lornage,X. etal. Recessive MYPN mutations cause
cap myopathy with occasional nemaline rods.
Ann.Neurol. 81, 467–473 (2017).
74. Miyatake,S. etal. Biallelic mutations in MYPN,
encoding myopalladin, are associated with childhood-
onset, slowly progressive nemaline myopathy.
Am.J.Hum. Genet. 100, 169–178 (2017).
75. Malfatti,E. etal. A premature stop codon in MYO18B
is associated with severe nemaline myopathy with
cardiomyopathy. J.Neuromuscul. Dis. 2, 219–227
(2015).
REVIEWS
164
|
MARCH 2018
|
VOLUME 14 www.nature.com/nrneurol
76. Domazetovska,A. etal. Intranuclear rod myopathy:
molecular pathogenesis and mechanisms of weakness.
Ann. Neurol. 62, 597–608 (2007).
77. Ryan,M.M. etal. Nemaline myopathy: a clinical study
of 143 cases. Ann. Neurol. 50, 312–320 (2001).
78. Feng,J.J. & Marston,S. Genotype–phenotype
correlations in ACTA1 mutations that cause congenital
myopathies. Neuromuscul. Disord. 19, 6–16 (2009).
79. Witting,N., Werlauff,U., Duno,M. & Vissing,J.
Prevalence and phenotypes of congenital myopathy
due to α-actin 1 gene mutations. Muscle Nerve 53,
388–393 (2016).
80. Jungbluth,H. etal. Mild phenotype of nemaline
myopathy with sleep hypoventilation due to a
mutation in the skeletal muscle α-actin (ACTA1) gene.
Neuromuscul. Disord. 11, 35–40 (2001).
81. Sambuughin,N. etal. KBTBD13 interacts with Cullin
3 to form a functional ubiquitin ligase. Biochem.
Biophys. Res. Commun. 421, 743–749 (2012).
82. Davidson,A.E. etal. Novel deletion of lysine 7
expands the clinical, histopathological and genetic
spectrum of TPM2-related myopathies. Brain 136,
508–521 (2013).
83. Jungbluth,H. etal. Magnetic resonance imaging of
muscle in nemaline myopathy. Neuromuscul. Disord.
14, 779–784 (2004).
84. Sato,I. etal. Congenital neuromuscular disease with
uniform type1 fiber and RYR1 mutation. Neurology
70, 114–122 (2008).
85. Muhammad,E. etal. Congenital myopathy is caused
by mutation of HACD1. Hum. Mol. Genet. 22,
5229–5236 (2013).
86. Maurer,M. etal. Centronuclear myopathy in Labrador
retrievers: a recent founder mutation in the PTPLA
gene has rapidly disseminated worldwide. PLoS ONE
7, e46408 (2012).
87. Walmsley,G.L. etal. Progressive structural defects
incanine centronuclear myopathy indicate a role for
HACD1 in maintaining skeletal muscle membrane
systems. Am. J.Pathol. 187, 441–456 (2017).
88. Clarke,N.F. etal. Mutations in TPM3 are a common
cause of congenital fiber type disproportion.
Ann.Neurol. 63, 329–337 (2008).
89. Munot,P. etal. Congenital fibre type disproportion
associated with mutations in the tropomyosin 3
(TPM3) gene mimicking congenital myasthenia.
Neuromuscul. Disord. 20, 796–800 (2010).
90. Clarke,N.F. etal. Recessive mutations in RYR1 are a
common cause of congenital fiber type disproportion.
Hum. Mutat. 31, E1544–E1550 (2010).
91. Laing,N.G. etal. Actin mutations are one cause of
congenital fibre type disproportion. Ann. Neurol. 56,
689–694 (2004).
92. Clarke,N.F. etal. SEPN1: associated with congenital
fiber-type disproportion and insulin resistance.
Ann.Neurol. 59, 546–552 (2006).
93. Lamont,P.J. etal. Novel mutations widen the
phenotypic spectrum of slow skeletal/β-cardiac myosin
(MYH7) distal myopathy. Hum. Mutat. 35, 868–879
(2014).
94. Vallat,J.M. etal. Coexistence of minicores, cores, and
rods in the same muscle biopsy. A new example of
mixed congenital myopathy. Acta Neuropathol. 58,
229–232 (1982).
95. Schartner,V. etal. Dihydropyridine receptor (DHPR.
CACNA1S) congenital myopathy. Acta Neuropathol.
133, 517–533 (2017).
96. Monnier,N. etal. Presence of two different genetic
traits in malignant hyperthermia families: implication
for genetic analysis, diagnosis, and incidence of
malignant hyperthermia susceptibility. Anesthesiology
97, 1067–1074 (2002).
97. Jurkat-Rott,K. etal. A calcium channel mutation
causing hypokalemic periodic paralysis. Hum. Mol.
Genet. 3, 1415–1419 (1994).
98. O’Grady,G.L. etal. Variants in the oxidoreductase
PYROXD1 cause early-onset myopathy with
internalized nuclei and myofibrillar disorganization.
Am. J.Hum. Genet. 99, 1086–1105 (2016).
99. Tajsharghi,H. & Oldfors,A. Myosinopathies:
pathology and mechanisms. Acta Neuropathol. 125,
3–18 (2013).
100. Tajsharghi,H. etal. Human disease caused by loss of
fast IIa myosin heavy chain due to recessive MYH2
mutations. Brain 133, 1451–1459 (2010).
101. Martinsson,T. etal. Autosomal dominant myopathy:
missense mutation (Glu-706 Lys) in the myosin
heavy chain IIa gene. Proc. Natl Acad. Sci. USA 97,
14614–14619 (2000).
102. Willis,T. etal. A novel MYH2 mutation in family
members presenting with congenital myopathy,
ophthalmoplegia and facial weakness. J.Neurol. 263,
1427–1433 (2016).
103. Tsabari,R., Daum,H., Kerem,E., Fellig,Y. & Dor,T.
Congenital myopathy due to myosin heavy chain 2
mutation presenting as chronic aspiration pneumonia
in infancy. Neuromuscul. Disord. 27, 947–950
(2017).
104. McMillin,M.J. etal. Mutations in ECEL1 cause distal
arthrogryposis type 5D. Am. J.Hum. Genet. 92,
150–156 (2013).
105. Dieterich,K. etal. The neuronal endopeptidase ECEL1
is associated with a distinct form of recessive distal
arthrogryposis. Hum. Mol. Genet. 22, 1483–1492
(2013).
106. Shaaban,S. etal. Expanding the phenotypic spectrum
of ECEL1-related congenital contracture syndromes.
Clin. Genet. 85, 562–567 (2014).
107. Todd,E.J. etal. Next generation sequencing in a large
cohort of patients presenting with neuromuscular
disease before or at birth. Orphanet J.Rare Dis. 10,
148 (2015).
108. Bayram,Y. etal. Molecular etiology of arthrogryposis in
multiple families of mostly Turkish origin. J.Clin. Invest.
126, 762–778 (2016).
109. Coste,B. etal. Gain-of-function mutations in the
mechanically activated ion channel PIEZO2 cause a
subtype of distal arthrogryposis. Proc. Natl Acad. Sci.
USA 110, 4667–4672 (2013).
110 . Zaharieva,I.T. etal. Loss-of-function mutations in
SCN4A cause severe foetal hypokinesia or ‘classical’
congenital myopathy. Brain 139, 674–691 (2016).
111. Singh,R.R. etal. Mutations in SCN4A: a rare but
treatable cause of recurrent life-threatening
laryngospasm. Pediatrics 134, e1447–e1450 (2014).
112 . Bharucha-Goebel,D.X. etal. Severe congenital
RYR1-associated myopathy: the expanding
clinicopathologic and genetic spectrum. Neurology
80, 1584–1589 (2013).
113 . Schessl,J. etal. MRI in DNM2-related centronuclear
myopathy: evidence for highly selective muscle
involvement. Neuromuscul. Disord. 17, 28–32
(2007).
114 . Clarke,N.F. etal. Cap disease due to mutation of the
beta-tropomyosin gene (TPM2). Neuromuscul. Disord.
19, 348–351 (2009).
115 . Lehtokari,V.L. etal. Cap disease caused by
heterozygous deletion of the β-tropomyosin gene
TPM2. Neuromuscul. Disord. 17, 433–442 (2007).
116 . Sewry,C.A., Holton,J.L., Dick,D.J., Muntoni,F. &
Hanna,M.G. Zebra body myopathy is caused by a
mutation in the skeletal muscle actin gene (ACTA 1 ).
Neuromuscul. Disord. 25, 388–391 (2015).
117 . Lacruz,R.S. & Feske,S. Diseases caused by mutations
in ORAI1 and STIM1. Ann. NY Acad. Sci. 1356,
45–79 (2015).
118 . Gordon,C.P. & Litz,S. Multicore myopathy in a
patient with anhidrotic ectodermal dysplasia.
Can.J.Anaesth. 39, 966–968 (1992).
119 . Engel,A.G., Redhage,K.R., Tester,D.J.,
Ackerman,M.J. & Selcen,D. Congenital myopathy
associated with the triadin knockout syndrome.
Neurology 88, 1153–1156 (2017).
120. Altmann,H.M. etal. Homozygous/compound
heterozygous triadin mutations associated with
autosomal-recessive long-QT syndrome and pediatric
sudden cardiac arrest: elucidation of the triadin
knockout syndrome. Circulation 131, 2051–2060
(2015).
121. Olive,M. etal. New cardiac and skeletal protein
aggregate myopathy associated with combined
MuRF1 and MuRF3 mutations. Hum. Mol. Genet. 24,
6264 (2015).
122. Zhang,L., Kelley,J., Schmeisser,G., Kobayashi,Y.M.
& Jones,L.R. Complex formation between junctin,
triadin, calsequestrin, and the ryanodine receptor.
Proteins of the cardiac junctional sarcoplasmic
reticulum membrane. J.Biol. Chem. 272,
23389–23397 (1997).
123. Park,H. etal. Comparing skeletal and cardiac
calsequestrin structures and their calcium binding:
aproposed mechanism for coupled calcium binding
and protein polymerization. J.Biol. Chem. 279,
18026–18033 (2004).
124. Costello,B. etal. Characterization of the junctional
face membrane from terminal cisternae of
sarcoplasmic reticulum. J.Cell Biol. 103, 741–753
(1986).
125. Treves,S. etal. Minor sarcoplasmic reticulum
membrane components that modulate excitation–
contraction coupling in striated muscles. J.Physiol.
587, 3071–3079 (2009).
126. Rios,E. & Gyorke,S. Calsequestrin, triadin and more:
the molecules that modulate calcium release in cardiac
and skeletal muscle. J.Physiol. 587, 3069–3070
(2009).
127. Guo,W. & Campbell,K.P. Association of triadin with
the ryanodine receptor and calsequestrin in the lumen
of the sarcoplasmic reticulum. J.Biol. Chem. 270,
9027–9030 (1995).
128. Wium,E., Dulhunty,A.F. & Beard,N.A. Three
residues in the luminal domain of triadin impact on
Trisk 95 activation of skeletal muscle ryanodine
receptors. Pflugers Arch. 468, 1985–1994 (2016).
129. Caswell,A.H., Motoike,H.K., Fan,H. & Brandt,N.R.
Location of ryanodine receptor binding site on
skeletal muscle triadin. Biochemistry 38, 90–97
(1999).
130. Groh,S. etal. Functional interaction of the
cytoplasmic domain of triadin with the skeletal
ryanodine receptor. J.Biol. Chem. 274,
12278–12283 (1999).
131. Goonasekera,S.A. etal. Triadin binding to the
C-terminal luminal loop of the ryanodine receptor is
important for skeletal muscle excitation contraction
coupling. J.Gen. Physiol. 130, 365–378 (2007).
132. Gordon,A.M., Homsher,E. & Regnier,M. Regulation
of contraction in striated muscle. Physiol. Rev. 80,
853–924 (2000).
133. Abu-Abed,M., Mal,T.K., Kainosho,M.,
MacLennan,D.H. & Ikura,M. Characterization of the
ATP-binding domain of the sarco(endo)plasmic
reticulum Ca
2+
-ATPase: probing nucleotide binding by
multidimensional NMR. Biochemistry 41, 1156–1164
(2002).
134. MacLennan,D.H., Asahi,M. & Tupling,A.R. The
regulation of SERCA-type pumps by phospholamban
and sarcolipin. Ann. NY Acad. Sci. 986, 472–480
(2003).
135. MacLennan,D.H. & Kranias,E.G. Phospholamban:
acrucial regulator of cardiac contractility. Nat. Rev.
Mol. Cell. Biol. 4, 566–577 (2003).
136. Asahi,M. etal. Sarcolipin regulates sarco(endo)
2+
-ATPase (SERCA) by binding to
transmembrane helices alone or in association with
phospholamban. Proc. Natl Acad. Sci. USA 100,
5040–5045 (2003).
137. Kurebayashi,N. & Ogawa,Y. Depletion of Ca
2+
in the
sarcoplasmic reticulum stimulates Ca
2+
entry into
mouse skeletal muscle fibres. J.Physiol. 533,
185–199 (2001).
138. Cherednichenko,G. etal. Conformational activation
ofCa
2+
entry by depolarization of skeletal myotubes.
Proc. Natl Acad. Sci. USA 101, 15793–15798
(2004).
139. Launikonis,B.S. & Rios,E. Store-operated Ca
2+
entry
during intracellular Ca
2+
release in mammalian
skeletal muscle. J.Physiol. 583, 81–97 (2007).
140. Stiber,J. etal. STIM1 signalling controls store-
operated calcium entry required for development and
contractile function in skeletal muscle. Nat. Cell Biol.
10, 688–697 (2008).
141. Peinelt,C. etal. Amplification of CRAC current by
STIM1 and CRACM1 (Orai1). Nat. Cell Biol. 8,
771–773 (2006).
142. Treves,S., Jungbluth,H., Muntoni,F. & Zorzato,F.
Congenital muscle disorders with cores: the ryanodine
receptor calcium channel paradigm. Curr. Opin.
Pharmacol. 8, 319–326 (2008).
143. Hwang,J.H., Zorzato,F., Clarke,N.F. & Treves,S.
Mapping domains and mutations on the skeletal
muscle ryanodine receptor channel. Trends Mol. Med.
18, 644–657 (2012).
144. Maclennan,D.H. & Zvaritch,E. Mechanistic models
for muscle diseases and disorders originating in the
sarcoplasmic reticulum. Biochim. Biophys. Acta 1813,
948–964 (2011).
145. Hirata,H. etal. Zebrafish relatively relaxed mutants
have a ryanodine receptor defect, show slow
swimming and provide a model of multi-minicore
disease. Development 134, 2771–2781 (2007).
146. Zhou,H. etal. RyR1 deficiency in congenital
myopathies disrupts excitation–contraction coupling.
Hum. Mutat. 34, 986–996 (2013).
147. Zhou,H. etal. Epigenetic allele silencing unveils
recessive RYR1 mutations in core myopathies.
Am.J.Hum. Genet. 79, 859–868 (2006).
148. Ducreux,S. etal. Functional properties of ryanodine
receptors carrying three amino acid substitutions
identified in patients affected by multi-minicore
disease and central core disease, expressed in
immortalized lymphocytes. Biochem. J. 395,
259–266 (2006).
REVIEWS
NATURE REVIEWS
|
NEUROLOGY VOLUME 14
|
MARCH 2018
|
165
149. Nelson,B.R. etal. Skeletal muscle-specific T-tubule
protein STAC3 mediates voltage-induced Ca
2+
release
and contractility. Proc. Natl Acad. Sci. USA 110,
11881–11886 (2013).
150. Polster,A., Nelson,B.R., Olson,E.N. & Beam,K.G.
Stac3 has a direct role in skeletal muscle-type
excitation–contraction coupling that is disrupted by a
myopathy-causing mutation. Proc. Natl Acad. Sci. USA
113 , 10986–10991 (2016).
151. Bohm,J. etal. Constitutive activation of the calcium
sensor STIM1 causes tubular-aggregate myopathy.
Am. J.Hum. Genet. 92, 271–278 (2013).
152. Volkers,M. etal. Orai1 deficiency leads to heart
failure and skeletal myopathy in zebrafish. J.Cell Sci.
125, 287–294 (2012).
153. Jurynec,M.J. etal. Selenoprotein N is required for
ryanodine receptor calcium release channel activity in
human and zebrafish muscle. Proc. Natl Acad. Sci.
USA 105, 12485–12490 (2008).
154. Arbogast,S. etal. Oxidative stress in SEPN1-related
myopathy: from pathophysiology to treatment.
Ann.Neurol. 65, 677–686 (2009).
155. Jungbluth,H. & Gautel,M. Pathogenic mechanisms in
centronuclear myopathies. Front. Aging Neurosci. 6,
339 (2014).
156. Cowling,B.S., Toussaint,A., Muller,J. & Laporte,J.
Defective membrane remodeling in neuromuscular
diseases: insights from animal models. PLoS Genet. 8,
e1002595 (2012).
157. Bachmann,C. etal. Cellular, biochemical and
molecular changes in muscles from patients with
X-linked myotubular myopathy due to MTM1
mutations. Hum. Mol. Genet. 26, 320–332 (2017).
158. Wallgren-Pettersson,C., Sewry,C.A., Nowak,K.J.
&Laing,N.G. Nemaline myopathies. Sem. Pediatr.
Neurol. 18, 230–238 (2011).
159. Ravenscroft,G. etal. Mouse models of dominant
ACTA 1 disease recapitulate human disease and
provide insight into therapies. Brain 134, 1101–1115
(2011).
160. Ravenscroft,G. etal. Actin nemaline myopathy mouse
reproduces disease, suggests other actin disease
phenotypes and provides cautionary note on muscle
transgene expression. PLoS ONE 6, e28699 (2011).
161. Jain,R.K. etal. Nemaline myopathy with stiffness
andhypertonia associated with an ACTA1 mutation.
Neurology 78, 1100–1103 (2012).
162. Donkervoort,S. etal. TPM3 deletions cause a
hypercontractile congenital muscle stiffness
phenotype. Ann. Neurol. 78, 982–994 (2015).
163. Ochala,J. etal. Disrupted myosin cross-bridge cycling
kinetics triggers muscle weakness in nebulin-related
myopathy. FASEB J. 25, 1903–1913 (2011).
164. Marttila,M. etal. Nebulin interactions with actin and
tropomyosin are altered by disease-causing mutations.
Skelet. Muscle 4, 15 (2014).
165. de Winter,J.M. etal. Mutation-specific effects on thin
filament length in thin filament myopathy. Ann. Neurol.
9, 959–969 (2016).
166. Ajima,R. etal. Deficiency of Myo18B in mice results
in embryonic lethality with cardiac myofibrillar
aberrations. Genes Cells 13, 987–999 (2008).
167. Gurung,R. etal. A zebrafish model for a human
myopathy associated with mutation of the
unconventional myosin MYO18B. Genetics 205,
725–735 (2017).
168. Gupta,V.A. & Beggs,A.H. Kelch proteins: emerging
roles in skeletal muscle development and diseases.
Skelet. Muscle 4, 11 (2014).
169. Garg,A. etal. KLHL40 deficiency destabilizes thin
filament proteins and promotes nemaline myopathy.
J.Clin. Invest. 124, 3529–3539 (2014).
170. Castets,P. etal. Satellite cell loss and impaired muscle
regeneration in selenoprotein N deficiency. Hum. Mol.
Genet. 20, 694–704 (2011).
171. Castets,P. etal. Selenoprotein N is dynamically
expressed during mouse development and detected
early in muscle precursors. BMC Dev. Biol. 9, 46
(2009).
172. Beggs,A.H. etal. MTM1 mutation associated with
X-linked myotubular myopathy in Labrador Retrievers.
Proc. Natl Acad. Sci. USA 107, 14697–14702
(2010).
173. Dowling,J.J., Low,S.E., Busta,A.S. & Feldman,E.L.
Zebrafish MTMR14 is required for excitation–
contraction coupling, developmental motor function
and the regulation of autophagy. Hum. Mol. Genet.
19, 2668–2681 (2010).
174. Fetalvero,K.M. etal. Defective autophagy and
mTORC1 signaling in myotubularin null mice.
Mol.Cell. Biol. 33, 98–110 (2013).
175. Al-Qusairi,L. etal. Lack of myotubularin (MTM1)
leads to muscle hypotrophy through unbalanced
regulation of the autophagy and ubiquitin–
proteasome pathways. FASEB J. 27, 3384–3394
(2013).
176. Durieux,A.C. etal. A centronuclear myopathy–
dynamin 2 mutation impairs autophagy in mice. Traffic
13, 869–879 (2012).
177. Sarparanta,J. etal. Interactions with M-band titin and
calpain 3 link myospryn (CMYA5) to tibial and limb-
girdle muscular dystrophies. J.Biol. Chem. 285,
30304–30315 (2010).
178. McClelland,V. etal. Vici syndrome associated with
sensorineural hearing loss and evidence of
neuromuscular involvement on muscle biopsy.
Am.J.Med. Genet. A 152A, 741–747 (2010).
179. Byrne,S. etal. EPG5-related Vici syndrome: a
paradigm of neurodevelopmental disorders with
defective autophagy. Brain 139, 765–781 (2016).
180. Rokach,O. etal. Epigenetic changes as a common
trigger of muscle weakness in congenital myopathies.
Hum. Mol. Genet. 24, 4636–4647 (2015).
181. North,K.N. etal. Approach to the diagnosis of
congenital myopathies. Neuromuscul. Disord. 24,
97–116 (2014).
182. Hacohen,Y. etal. Fetal acetylcholine receptor
inactivation syndrome: a myopathy due to maternal
antibodies. Neurol. Neuroimmunol. Neuroinflamm. 2,
e57 (2015).
183. Kinali,M. etal. Congenital myasthenic syndromes in
childhood: diagnostic and management challenges.
J.Neuroimmunol. 201–202, 6–12 (2008).
184. Bonnemann,C.G. etal. Diagnostic approach to the
congenital muscular dystrophies. Neuromuscul.
Disord. 24, 289–311 (2014).
185. Selcen,D. Myofibrillar myopathies. Neuromuscul.
Disord. 21, 161–171 (2011).
186. Nishino,I. Autophagic vacuolar myopathy. Semin.
Pediatr. Neurol. 13, 90–95 (2006).
187. Wang,C.H. etal. Consensus statement on standard of
care for congenital myopathies. J.Child Neurol. 27,
363–382 (2012).
188. Jungbluth,H., Ochala,J., Treves,S. & Gautel,M.
Current and future therapeutic approaches to the
congenital myopathies. Semin. Cell Dev. Biol. 64,
191–200 (2017).
189. Childers,M.K. etal. Gene therapy prolongs survival
and restores function in murine and canine models of
myotubular myopathy. Sci. Transl Med. 6, 220ra10
(2014).
190. Rendu,J. etal. Exon skipping as a therapeutic
strategy applied to an RYR1 mutation with pseudo-
exon inclusion causing a severe core myopathy. Hum.
Gene Ther. 24, 702–713 (2013).
191. Monnier,N. etal. A homozygous splicing mutation
causing a depletion of skeletal muscle RYR1 is
associated with multi-minicore disease congenital
myopathy with ophthalmoplegia. Hum. Mol. Genet.
12, 1171–1178 (2003).
192. Barton-Davis,E.R., Cordier,L., Shoturma,D.I.,
Leland,S.E. & Sweeney,H.L. Aminoglycoside
antibiotics restore dystrophin function to skeletal
muscles of mdx mice. J.Clin. Invest. 104, 375–381
(1999).
193. MacArthur,D.G. & Lek,M. The uncertain road
towards genomic medicine. Trends Genet. 28,
303–305 (2012).
194. Cowling,B.S. etal. Reducing dynamin 2 expression
rescues X-linked centronuclear myopathy. J.Clin.
Invest. 124, 1350–1363 (2014).
195. Sabha,N. etal. PIK3C2B inhibition improves function
and prolongs survival in myotubular myopathy animal
models. J.Clin. Invest. 126, 3613–3625 (2016).
196. Ravenscroft,G. etal. Cardiac α-actin over-expression
therapy in dominant ACTA1 disease. Hum. Mol. Genet.
22, 3987–3997 (2013).
197. Nowak,K.J. etal. Nemaline myopathy caused by
absence of α-skeletal muscle actin. Ann. Neurol. 61,
175–184 (2007).
198. Lawlor,M.W. etal. Enzyme replacement therapy
rescues weakness and improves muscle pathology in
mice with X-linked myotubular myopathy. Hum. Mol.
Genet. 22, 1525–1538 (2013).
199. Fruen,B.R., Mickelson,J.R. & Louis,C.F. Dantrolene
inhibition of sarcoplasmic reticulum Ca
2+
release by
direct and specific action at skeletal muscle ryanodine
receptors. J.Biol. Chem. 272, 26965–26971
(1997).
200. Timmins,M.A. etal. Malignant hyperthermia testing
in probands without adverse anesthetic reaction.
Anesthesiology 123, 548–556 (2015).
201. Michalek-Sauberer,A. & Gilly,H. Prophylactic use of
dantrolene in a patient with central core disease.
Anesth. Analg. 86, 915–916 (1998).
202. Jungbluth,H., Dowling,J.J., Ferreiro,A. & Muntoni,F.
217th ENMC International Workshop: RYR1-related
myopathies, Naarden, The Netherlands,
29–31January 2016. Neuromuscul. Disord. 26,
624–633 (2016).
203. Andersson,D.C. & Marks,A.R. Fixing ryanodine
receptor Ca leak — a novel therapeutic strategy for
contractile failure in heart and skeletal muscle. Drug
Discov. Today Dis. Mech. 7, e151–e157 (2010).
204. Marks,A.R. Calcium cycling proteins and heart
failure: mechanisms and therapeutics. J.Clin. Invest.
123, 46–52 (2013).
205. Pold,R. etal. Long-term AICAR administration and
exercise prevents diabetes in ZDF rats. Diabetes 54,
928–934 (2005).
206. Lanner,J.T. etal. AICAR prevents heat-induced
sudden death in RyR1 mutant mice independent
ofAMPK activation. Nat. Med. 18, 244–251
(2012).
207. de Winter,J.M. etal. Troponin activator augments
muscle force in nemaline myopathy patients with
nebulin mutations. J.Med. Genet. 50, 383–392
(2013).
208. de Winter,J.M. etal. Effect of levosimendan on the
contractility of muscle fibers from nemaline myopathy
patients with mutations in the nebulin gene. Skelet.
Muscle 5, 12 (2015).
209. Amthor,H. & Hoogaars,W.M. Interference with
myostatin/ActRIIB signaling as a therapeutic strategy
for Duchenne muscular dystrophy. Curr. Gene Ther.
12, 245–259 (2012).
210. Durham,W.J. etal. RyR1 S-nitrosylation underlies
environmental heat stroke and sudden death in
Y522S RyR1 knockin mice. Cell 133, 53–65
(2008).
211 . Dowling,J.J. etal. Oxidative stress and successful
antioxidant treatment in models of RYR1-related
myopathy. Brain 135, 1115–1127 (2012).
212. Natera-de Benito,D. etal. KLHL40-related nemaline
myopathy with a sustained, positive response to
treatment with acetylcholinesterase inhibitors.
J.Neurol. 263, 517–523 (2016).
213. Robb,S.A. etal. Impaired neuromuscular
transmission and response to acetylcholinesterase
inhibitors in centronuclear myopathies. Neuromuscul.
Disord. 21, 379–386 (2011).
214. Gibbs,E.M. etal. Neuromuscular junction
abnormalities in DNM2-related centronuclear
myopathy. J.Mol. Med. 91, 727–737 (2013).
215. Dowling,J.J. etal. Myotubular myopathy and the
neuromuscular junction: a novel therapeutic approach
from mouse models. Dis. Model. Mech. 5, 852–859
(2012).
216. Messina,S. etal. Pilot trial of salbutamol in central
core and multi-minicore diseases. Neuropediatrics 35,
262–266 (2004).
217. Schreuder,L.T. etal. Successful use of albuterol in a
patient with central core disease and mitochondrial
dysfunction. J.Inherit. Metab. Dis. 33, S205–209
(2010).
218. Jungbluth,H., Dowling,J.J., Ferreiro,A. & Muntoni,F.
182nd ENMC International Workshop: RYR1-related
myopathies, 15–17th April 2011, Naarden, The
Netherlands. Neuromuscul. Disord. 22, 453–462
(2012).
219. Nguyen,M.A. etal. Hypertrophy and dietary tyrosine
ameliorate the phenotypes of a mouse model of
severe nemaline myopathy. Brain 134, 3516–3529
(2011).
220. Ryan,M.M. etal. Dietary L-tyrosine supplementation
in nemaline myopathy. J.Child Neurol. 23, 609–613
(2008).
221. Winter,L. etal. Chemical chaperone ameliorates
pathological protein aggregation in plectin-deficient
muscle. J.Clin. Invest. 124, 1144–1157 (2014).
222. Kusaczuk,M., Bartoszewicz,M. & Cechowska-
Pasko,M. Phenylbutyric acid: simple structure —
multiple effects. Curr. Pharm. Des. 21, 2147–2166
(2015).
223. Cuadrado-Tejedor,M., Ricobaraza,A.L., Torrijo,R.,
Franco,R. & Garcia-Osta,A. Phenylbutyrate is a
multifaceted drug that exerts neuroprotective effects
and reverses the Alzheimer’s disease-like phenotype of
a commonly used mouse model. Curr. Pharm. Des. 19,
5076–5084 (2013).
224. Lee,C.S. etal. A chemical chaperone improves muscle
function in mice with a RyR1 mutation. Nat. Commun.
8, 14659 (2017).
REVIEWS
166
|
MARCH 2018
|
VOLUME 14 www.nature.com/nrneurol
225. Vega,H., Agellon,L.B. & Michalak,M. The rise of
proteostasis promoters. IUBMB Life 68, 943–954
(2016).
226. Yuste-Checa,P. etal. Pharmacological chaperoning: a
potential treatment for PMM2-CDG. Hum. Mutat. 38,
160–168 (2017).
227. Kim,E., etal. Characterization of human cardiac
calsequestrin and its deleterious mutants. J. Mol. Biol.
373, 1047–1057 (2007).
228. Tang,L., etal. Structural basis for Ca
2+
selectivity of a
voltage-gated calcium channel. Nature 505, 56–61
(2014).
229. Zamoon, J., Mascioni, A., Thomas, D.D. & Veglia, G.
NMR solution structure and topological orientation of
monomeric phospholamban in dodecylphosphocholine
micelles. Biophys. J. 85, 2589–2598 (2003).
230. Efremov, R.G., Leitner,A., Aebersold,R. & Raunser,S.
Architecture and conformational switch mechanism
ofthe ryanodine receptor. Nature 517, 39–43
(2015).
231. Toyoshima,C., Nakasako,M., Nomura,H. &
Ogawa,H. Crystal structure of the calcium pump of
sarcoplasmic reticulum at 2.6 Å resolution. Nature
405, 647–655 (2000).
Author contributions
H.J., S.T., F.Z., J.O., C.S., M.G. and F.M. researched data for
the article. All authors made substantial contributions to dis-
cussion of the content, wrote the article and reviewed and/or
edited the manuscript before submission.
Competing interests statement
The authors declare no competing interests.
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional
claims in published maps and institutional affiliations.
REVIEWS
NATURE REVIEWS
|
NEUROLOGY VOLUME 14
|
MARCH 2018
|
167