Sei sulla pagina 1di 23

doi:10.

1093/brain/awu368 BRAIN 2015: 138; 246–268 | 246

REVIEW ARTICLE
Pathophysiological concepts in the congenital
myopathies: blurring the boundaries,
sharpening the focus
Gianina Ravenscroft,1 Nigel G. Laing1 and Carsten G. Bönnemann2

The congenital myopathies are a diverse group of genetic skeletal muscle diseases, which typically present at birth or in early
infancy. There are multiple modes of inheritance and degrees of severity (ranging from foetal akinesia, through lethality in the
newborn period to milder early and later onset cases). Classically, the congenital myopathies are defined by skeletal muscle
dysfunction and a non-dystrophic muscle biopsy with the presence of one or more characteristic histological features. However,
mutations in multiple different genes can cause the same pathology and mutations in the same gene can cause multiple different
pathologies. This is becoming ever more apparent now that, with the increasing use of next generation sequencing, a genetic
diagnosis is achieved for a greater number of patients. Thus, considerable genetic and pathological overlap is emerging, blurring
the classically established boundaries. At the same time, some of the pathophysiological concepts underlying the congenital
myopathies are moving into sharper focus. Here we explore whether our emerging understanding of disease pathogenesis and
underlying pathophysiological mechanisms, rather than a strictly gene-centric approach, will provide grounds for a different and
perhaps complementary grouping of the congenital myopathies, that at the same time could help instil the development of shared
potential therapeutic approaches. Stemming from recent advances in the congenital myopathy field, five key pathophysiology
themes have emerged: defects in (i) sarcolemmal and intracellular membrane remodelling and excitation-contraction coupling;
(ii) mitochondrial distribution and function; (iii) myofibrillar force generation; (iv) atrophy; and (v) autophagy. Based on numerous
emerging lines of evidence from recent studies in cell lines and patient tissues, mouse models and zebrafish highlighting these
unifying pathophysiological themes, here we review the congenital myopathies in relation to these emerging pathophysiological
concepts, highlighting both areas of overlap between established entities, as well as areas of distinction within single gene disorders.

1 Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, Western
Australia, Australia
2 National Institute of Neurological Disorders and Stroke/NIH, Porter Neuroscience Research Centre, Bethesda, MD, USA
Correspondence to: Carsten G. Bönnemann, MD,
Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch
National Institute of Neurological Disorders and Stroke/NIH
Porter Neuroscience Research Centre 35 Convent Drive, Bldg 35, Room 2A-116
Bethesda, MD 20892-3705
E-mail: carsten.bonnemann@nih.gov

Keywords: congenital myopathy; pathophysiology; mitochondria; membrane remodelling; force generation; atrophy; autophagy;
triad; excitation-contraction coupling
Abbreviations: CNM = centronuclear myopathy; DHPR = dihydropyridine receptors; KI/KO = knock-in/knock-out;
NMJ = neuromuscular junction; SR = sarcoplasmic reticulum

Received May 6, 2014. Revised November 21, 2014. Accepted November 22, 2014. Advance Access publication December 31, 2014
Published by Oxford University Press on behalf of the Guarantors of Brain 2014. This work is written by US Government employees and is in the public domain in the US.
Pathophysiology of congenital myopathies BRAIN 2015: 138; 246–268 | 247

heterogeneity, through the continuous identification of


Introduction novel disease genes, has led to an increased blurring of
The congenital myopathies are clinically defined by hypo- the established diagnostic boundaries between the subsets
tonia and skeletal muscle weakness and pathologically by of the classic congenital myopathies. Such novel genes pro-
the presence of one or more histopathological or ultrastruc- viding new mechanistic aspects have included CCDC78 in
tural hallmarks on muscle biopsy (North, 2011; Romero a family with autosomal dominant CNM with cores
and Clarke, 2013). In extremely rare instances, congenital (Majczenko et al., 2012), HACD1 (now known as
myopathy may also be associated with hypertonia as will PTPLA) in a family with a non-specific autosomal recessive
be discussed below (Jain et al., 2012; C. G. Bönnemann congenital myopathy (Muhammad et al., 2013), STAC3 in
et al., unpublished data). There are three broad histopatho- Native American myopathy (Horstick et al., 2013), STIM1
logical subtypes within the classical congenital myopathies: in tubular aggregate myopathy (although this is not strictly
core myopathies characterized by the presence of myofibres a congenital myopathy) (Bohm et al., 2013a) but also TTN
with foci devoid of oxidative enzymes (Jungbluth et al., mutation in patients presenting with myopathies character-
2011); centronuclear myopathies (CNMs) defined by the ized by internal nuclei and/or cores (Ceyhan-Birsoy et al.,
presence of internally located myonuclei (Romero and 2013; Chauveau et al., 2014). In this review we have there-
fore focused on the emerging pathophysiological themes
Bitoun, 2011) (which are now recognized as one of the
across these entities, under the hypothesis that increased
most common type of congenital myopathy, due to the
understanding of these mechanistic aspects of disease will
relatively high prevalence of RYR1 mutations that can
not only help classification, but will ultimately facilitate the
also be associated with this histological phenotype)
preclinical development of therapeutic approaches for these
(Amburgey et al., 2011; Maggi et al., 2013); and nemaline
diseases (see Fig. 1 for a schematic guide to the cellular
myopathy, defined by the presence of electron dense nema-
context of the pathophysiological concepts discussed in
line bodies/rods within myofibres (Wallgren-Pettersson
the review and table 1 for a correlation of individual
et al., 2011). In addition, there are a number of other
genetic entities and associated pathophysiological themes).
structural abnormalities that can be observed in the con-
Given the focus of this review, we are not redescribing the
genital myopathies including actin aggregates, caps, cylin-
clinical entities but refer the reader to recent excellent re-
drical spirals, dystrophic-like features, fibre-type
views (Nance et al., 2012; Maggi et al., 2013; Romero
disproportion, intranuclear rods, lobulated myofibres, neck-
and Clarke, 2013).
lace myofibres and zebra bodies (Clarke, 2011; Goebel and
Blaschek, 2011). There are also frequently cases that pre-
sent with mixed morphologies, such as cores and nemaline
bodies (Romero et al., 2009; Olive et al., 2010) or cores
Membrane remodelling
and internal nuclei (Wilmshurst et al., 2010). defects
It has been increasingly recognized that the histological
features observed within a muscle biopsy may represent
manifestations along spectra of possible abnormalities,
Membrane remodelling and the
rather than discrete pathological entities (Sewry et al., phosphoinositide phosphatases
2008; Nance et al., 2012). For example central nuclei are Membrane remodelling is crucial to a number of biological
classically associated with mutations in BIN1, DNM2, processes including exocytosis, intracellular transport, syn-
MTM1, and SPEG however, more recently, prominent aptic vesicle function, autophagy (see separate discussion
central nuclei have been reported in cases resulting from below), and membrane repair. These processes involve pro-
mutations in CFL2 (Ockeloen et al., 2012), KBTBD13 teins that regulate lipids; they act as lipid adaptor molecules
(Olive et al., 2010), RYR1 (Wilmshurst et al., 2010) and and function to regulate cytoskeletal organization.
TTN (Ceyhan-Birsoy et al., 2013; Palmio et al., 2014). On Strikingly, a number of genes found to underlie congenital
the flip side, although congenital myopathies have trad- myopathies characterized by the presence of internal myonu-
itionally been classified purely by their defining histopath- clei, are involved in such membrane remodelling [amphi-
ology, with the discovery of the underlying genetic cause physin 2/bridging integrator 1, BIN1 (MIM 601248);
for an increasing number of patients, the field has also dynamin 2, DNM2 (MIM 602378); myotubularin, MTM1
begun in parallel to classify patients based on the causative (MIM 300415) and myotubularin-related protein 14,
disease gene, such that there is now, for instance, reference MTMR14 (MIM 611089)]; (reviewed in De Matteis and
to ACTA1-related myopathies (Nowak et al., 2013), Luini, 2011; Cowling et al., 2012) and most recently, striated
RYR1-related myopathies (Klein et al., 2012; Amburgey preferentially expressed gene, SPEG (Agrawal et al., 2014).
et al., 2013; Bharucha-Goebel et al., 2013) and SEPN1- Myotubularin (MTM1), found to be the cause of X-
related myopathies (Schara et al., 2008; Scoto et al., linked myotubular myopathy (XLMTM, MIM 310400),
2011). Each of these gene-associated spectra has continued is the canonical member of a family of phosphoinositide
to expand with multiple possible morphological appear- phosphatases. The mammalian myotubularin family com-
ances per gene, which together with ever-greater genetic prises MTM1 itself and 14 MTM1-related proteins
248 | BRAIN 2015: 138; 246–268 G. Ravenscroft et al.

Figure 1 Schematic representation of a muscle cell placing the general pathophysiological concepts discussed in this review
into the cellular context. (1) Sarcolemmal and intracellular membrane remodelling and excitation-contraction coupling; (2) mitochondrial
distribution and function; (3) myofibrillar force generation; (4) atrophy; and (5) autophagy.

(MTMR1–14). Phosphoinositide phosphatases are crucial disturbance of membrane metabolism is still not completely
to the maintenance of phosphoinositides (PIs) and via the understood; however, studies in animal models are begin-
regulation of these metabolites, are central to membrane ning to shed light in this area, highlighting several potential
trafficking and signalling (Di Paolo and De Camilli, 2006; mechanisms that are addressed in the following sections.
Nicot and Laporte, 2008). The key metabolites regulated
by myotubularins are phosphatidylinositol 3-phosphate Excitation-contraction coupling, transverse-tubule
(PI3P), which acts as a membrane address marker and and sarcoplasmic reticulum defects
functions to recruit proteins to the membrane, and The precise regulation of intracellular Ca2+ is crucial for
PtdIns(3,5)P2 and PtdIns(5)P. MTM1 dephosphorylates normal myogenesis and skeletal muscle function.
phosphoinositides, including PI3P [produced by PI Excitation-contraction (E–C) coupling exquisitely modu-
3-kinase (PI3K) activity] and PtdIns(3,5)P2 [PI3,5P2; pro- lates sarcoplasmic Ca2+ levels during muscle contraction
duced by PI 5-kinase (PI5K) activity]. Mutations in and depends on a number of specialized membranes and
MTMR14 have been associated with myopathy in fish structural components including components of the triad,
(Dowling et al., 2010) and possibly CNM in man (Tosch namely T tubules and the voltage-sensing dihydropyridine
et al., 2006), although the evidence for the latter remains receptors (DHPR) in the T tubule membrane, and the
incomplete. It is worth noting that mutation of MTMR2 sarcoplasmic reticulum (SR) and the Ca2+ release chan-
and MTMR13 are responsible for some cases of autosomal nels/ryanodine receptors (RYR1) in the sarcoplasmic reticu-
recessive Charcot–Marie–Tooth type hereditary motor and lum membrane. Structural defects of triads and abnormal
sensory neuropathies (CMT4B), which is of interest be- localization of both DHPR and RYR1 have been noted in
cause mutations in DNM2 are also seen in both CNM myofibres from patients with CNMs due to MTM1, BIN1
and in autosomal dominant CMT (CMTDIB) (De Matteis and CCDC78 (MIM 614666) mutations (Al-Qusairi et al.,
and Luini, 2011). How disrupted phosphoinositide regula- 2009; Dowling et al., 2009; Toussaint et al., 2011;
tion leads to neuromuscular disease via possible Majczenko et al., 2012). Similar defects have also been
Pathophysiology of congenital myopathies BRAIN 2015: 138; 246–268 | 249

Table 1 Summary of the pathophysiological features associated with particular congenital myopathy disease genes

Gene Membrane defects Mitochondrial and energy Myofibrillar force Atrophy Autophagy
metabolism abnormalities defects defects defects
T tubules/SR NMJ
BIN1 3
DNM2 3 3 3
MTM1 3 3 3 3 3
MTMR14 3 3 3
RYR1 3 3 3
ACTA1 3 3
CCDC78 3
KBTBD13 3
KLHL40 3
MEGF10 3
MYH7 3
NEB 3 3
TPM2 3
TPM3 3 3 3
SEPN1 3 3

observed and more closely investigated in animal models of reticulum networks, to severe disorganization of the T tu-
the various CNMs. bules and sarcoplasmic reticulum (Dowling et al., 2009).
The most well studied myotubularin model is the Mtm1
knock-out (KO) mouse. These mice develop a generalized
Myotubularin
and progressive myopathy from 4 weeks of age with
Studies of Drosophila myotubularin (mtm1) showed that
increased internalized nuclei leading to death at 6–14
mtm1 is central to endolysosomal function, cortical actin weeks of age (Buj-Bello et al., 2002). Analysis of Mtm1
remodelling and is also involved in integrin-mediated myo- null mouse muscle found that the sarcoplasmic reticulum
fibre attachment (Velichkova et al., 2010). Mtm1 ablation membranes were abnormally located within myofibres and
resulted in accumulation of integrin and PI3P on endo- that the sarcoplasmic reticulum was dilated (Amoasii et al.,
somes, suggesting that mtm1 is required for intracellular 2013). In order to study the SR/ER (endoplasmic reticulum)
integrin trafficking and recycling to the sarcolemma in isolation, Amoasii and colleagues (2013) examined the
(Ribeiro et al., 2011). The phenotype could be rescued endoplasmic reticulum network in myoblasts from Mtm1
by depletion of class II PI3K, confirming that the abnorm- null mice and MTM1 patients. The myoblasts lacked triads
alities observed are truly IP-related. and T tubules and showed defects in the endoplasmic
Knock-down of mtm1 in zebrafish resulted in delayed reticulum network, suggesting that an absence of MTM1
hatching and an impaired touch-evoked escape contraction resulted in sarcoplasmic reticulum defects (as the sarcoplas-
(Dowling et al., 2009). Muscle from the mtm1 morphants mic reticulum is a specialization of the endoplasmic reticu-
displayed centrally located and abnormally enlarged nuclei lum) independent of abnormalities of the T tubules and
and skeletal muscle hypotrophy, mimicking the human dis- triads. This suggests a general defect in the maintenance
ease. Interestingly, the levels of PI3P were elevated in the of various membranous muscle organelles. In vivo modu-
skeletal musculature of mtm1 morphants relative to con- lation of MTM1 lipid phosphatase activity in normal
trols (similar to the Drosophila studies), but they were not mouse muscle using viral overexpression of wild-type or
elevated in whole embryo extracts, suggesting that myotu- mutant (phosphatase inactive) MTM1, resulted in altered
bularin is the primary phosphoinositide phosphatase in PI3P levels specifically at the junctional sarcoplasmic reticu-
skeletal muscle (Dowling et al., 2009). The skeletal lum as well as altered sarcoplasmic reticulum remodelling.
muscle phenotype of the mtm1 morphants was rescued Importantly, this study suggests that dysregulation of the
by delivery of MTMR1 and MTMR2, indicating that MTM1/PI3P pathway, leading to altered modulation of the
the primary defect in the morphants was due to a lack of sarcoplasmic reticulum structure, may represent a crucial
compensation by other phosphoinositide phosphatases mechanism underlying disease pathogenesis in myotubular
(Dowling et al., 2009). In zebrafish (Dowling et al., myopathy.
2009) and mice (Amoasii et al., 2013), myotubularin local- This same group also showed that the phenotype of
izes to the T tubules as evidenced by co-localization with the Mtm1 null mouse could be partially rescued by
antibodies to the DHPR. The perinuclear compartments of adeno-associated viral delivery of the MTM1Cys375Ser or
mtm1 morphant myofibres displayed abnormal tubule-like MTM1Ser376Asn phosphatase-dead mutants, including a sig-
structures, ranging from dilated T tubule and sarcoplasmic nificant increase in muscle force generation, suggesting that
250 | BRAIN 2015: 138; 246–268 G. Ravenscroft et al.

many features of the disease are not purely due to the with functionally important abnormalities of triadic struc-
enzymatic defect (muscle atrophy, myofibre hypotrophy, ture with consequences on excitation-contraction coupling.
abnormal organelle placement) (Amoasii et al., 2012).
Consistent with this is the consideration that MTM1 is MTMR14
quite ubiquitously expressed with not only skeletal muscle MTMR14 is a phosphatase that shares similar enzymatic
specific localization, whereas XL-CNM is largely (though activity to MTM1. Although two human MTMR14 vari-
not exclusively) a skeletal muscle disease suggesting that ants have been described, and were shown to reduce the
defects in interactions between MTM1 and muscle-specific enzymatic activity of MTMR14 (Tosch et al., 2006), they
proteins could play a major role in disease causation. The are still awaiting further confirmation as a direct cause of
most evident extra-phosphatase activity for MTM1 might CNM or a disease modifier. Of the two variants described,
be in protein–protein interactions and functioning as a scaf- one de novo MTMR14 missense variant (p.Y462C) was
fold for the intermediate filament network (desmin in par- identified in an isolated proband in combination with a
ticular) (Hnia et al., 2011); which is involved in proper de novo known DNM2 mutation (p.E368K), the
organelle placement (Amoasii et al., 2012). p.Y462C variant was also identified in a control sample.
The mouse model generated by knocking-in the human The second proband harboured a p.R336Q variant in
disease associated mutation p.Arg69Cys in Mtm1 is less MTMR14, this variant was also present in the unaffected
severe compared to the Mtm1 KO model, likely due to father (Tosch et al., 2006). Inactivation of Mtmr14 in mice
some residual activity of the full length mutant myotubu- (Shen et al., 2009; Hnia et al., 2012) and fish causes a
larin (Pierson et al., 2012). Mtm1 p.Arg69Cys knock-in myopathy characterized by excitation-contraction coupling
(KI) mice are asymptomatic at birth and show normal ini- defects but without the presence of central or abnormally
tial skeletal muscle development. However, by 2 months of located myonuclei. In zebrafish, knock-down of mtmr14
age muscle atrophy and weakness is detected. These mice resulted in a functional deficit (reduced touch-evoked
had a median survival age of 66 weeks and died suddenly escape responses and a lack of response to high frequency
and unexpectedly, consistent with cardiorespiratory failure. stimulation) without obvious histopathological abnormal-
Histologically, skeletal muscle showed many small myofi- ities (Dowling et al., 2010). Hnia et al. (2012) showed
bres, central nuclei, staining suggestive of necklace fibres that muscle from Mtmr14 KO mice displayed significantly
similar to the human pathology, and altered staining pat- swollen and elongated T tubules and suggested that these
terns with antibodies to RYR1 and DHPR. Reduced num- structural abnormalities might contribute to the defects in
bers of, and poorly formed, T tubules and triads were excitation-contraction coupling noted previously in
identified at the ultrastructural level (Pierson et al., Mtmr14 KO mice (Shen et al., 2009) and morphant fish
2012). These morphological abnormalities result in (Dowling et al., 2010).
excitation-contraction coupling defects, since for Mtm1
KO and p.Arg69Cys KI mice, electrically-mediated whole Amphiphysin 2 and dynamin 2
muscle-specific force was greatly reduced, whereas there Altered T-tubular and sarcoplasmic reticulum systems have
was no change between specific force generated by wild- also been noted due to mutation in DNM2 and BIN1,
type and Mtm1 chemically-skinned (in which T tubules and which are both known to underlie forms of centronuclear
triads are not functioning) myofibres (Lawlor et al., 2013). myopathy. Early studies showed that amphiphysin 2
Most recently, adeno-associated virus-mediated knock- (BIN1) was concentrated at the T tubules and was required
down of Mtm1 in adult mouse muscle resulted in for T tubule biogenesis. Expression of BIN1 in non-muscle
abnormally oriented T tubules that were more frequently cells resulted in tubular invaginations of the plasma mem-
appearing as longitudinal (L-) tubules (Joubert et al., 2013) brane (Lee et al., 2002).
highlighting that myotubularin is continuously required for The original study that showed that BIN1 mutations
adult skeletal muscle organelle maintenance. resulted in autosomal recessive-CNM, also showed that
Beggs and colleagues (2010) reported a Labrador missense mutations resulted in impaired tubule formation
Retriever line with a missense mutation in MTM1; the and pull-down assays showed that partial truncation of
affected male dogs displayed many of the clinical and histo- BIN1 abrogated the interaction with DNM2 (Nicot et al.,
logical hallmark features of human MTM1 related XL- 2007). The authors concluded that BIN1 mutation caused
CNM, presenting with progressive limb weakness leading CNM by interfering with remodelling of T tubules and that
to an inability to rise or walk without assistance between interaction between BIN1 and DNM2 is required for
10–19 weeks of age. Clinically, there was severe muscle muscle function and myonuclei positioning. Most recently,
atrophy. Histologically there were numerous small central- BIN1 has also been shown to interact directly with MTM1
ly-nucleated myofibres, resembling myotubes, necklace (Royer et al., 2013).
myofibres, abnormal localization of RYR1 and DHPR BIN1 is ubiquitously expressed and there are at least 12
and diminished and disturbed T tubule structures (Beggs different splice isoforms, those containing a phosphoinosi-
et al., 2010). Thus in both non-mammalian and mamma- tide domain are found exclusively in skeletal muscle
lian models (small and large) the pathophysiology of isoforms, which all contain exon 11 (Toussaint et al.,
MTM1-related disease is remarkably similar and consistent 2011). Bohm et al. (2013b) identified the first mutation
Pathophysiology of congenital myopathies BRAIN 2015: 138; 246–268 | 251

affecting the muscle-specific BIN1 exon 11 in a consan- so that reduction of DNM2 restores this balance in
guineous family presenting with a rapidly progressive and MTM1 deficiency. This approach could thus be beneficial
fatal CNM. Mutation of the same acceptor-splice site in in XLMTM.
exon 11 was identified to underlie canine inherited myop-
athy of Great Danes (IMGD). Cell-culture studies showed SPEG
that BIN1 constructs without exon 11 were unable to ini- Agrawal and colleagues (2014) identified SPEG as a novel
tiate membrane tubulation (Bohm et al., 2013b). Analysis binding partner of MTM1 in a yeast two-hybrid screen and
of patient and inherited myopathy of Great Danes muscle subsequently showed that it likely localized to the terminal
showed central nuclei, fibre atrophy and lobulation, abnor- cisternae of the sarcoplasmic reticulum. Recessive muta-
mal triads, accumulations of membranes and vacuoles, tions of SPEG were identified in patients with a severe
autophagic vacuoles and mitochondrial accumulations. form of CNM, two cases also had cardiomyopathy
Proteins of the triad and sarcoplasmic reticulum (RYR1, (Agrawal et al., 2014). Speg / mice display perinatal
DHPR, SERCA1) and membrane-trafficking proteins death due to dilated cardiomyopathy (Liu et al., 2009),
(MTM1, CAV3, DYSF) were also mislocalized in inherited evaluation of Speg / skeletal muscle revealed increased
myopathy of Great Danes muscle. central nuclei (Agrawal et al., 2014). While attractive to
DNM2-related dysfunction has been studied in zebrafish consider, it remains to be seen whether loss of SPEG
and mice. Zebrafish overexpressing the p.Ser619Leu substi- leads to defects in membrane remodelling and tubulation,
tution in DNM2 showed impaired motility, with smaller as occurs in MTM1-, BIN1- and DNM2-related CNM.
myofibres, and dilated sarcoplasmic reticulum (Gibbs
et al., 2013). More recently it was shown that the Triadic Ca2+ release channel: ryanodine receptor
DNM2 p.Ser619Leu zebrafish accumulate aberrant ves- The so-called ryanodine receptor (RYR1) is the major Ca2+
icle material and show defects of excitation-contraction release channel of the sarcoplasmic reticulum. It is located
coupling. Expression of the p.Ser619Leu mutant in at the triad, where it directly interacts with the voltage-
non-muscle cells resulted in defective BIN1-dependent tubu- sensitive dihydropyridine receptor in the T tubule. Via
lation (Gibbs et al., 2014). Thus it seems that an under- this connection it mediates excitation initiated Ca2+ release
lying mechanism in the CNMs is defective membrane into the sarcoplasm (Zorzato et al., 1990).
tubulation. Myopathies resulting from autosomal dominant RYR1
Studies of a KI mouse model of the most frequently (MIM 180901) mutations [including malignant hyperther-
occurring DNM2 mutation (p.Arg465Trp) revealed that mia susceptibility (MHS; MIM 145600), central core dis-
muscle from heterozygous KI mice contains increased tubu- ease (MIM 117000), multi-minicore disease, congenital
lar structures within the intermyofibrillar space and slightly fibre-type disproportion (CFTD), some subtypes of CNM
disorganized T tubule and sarcoplasmic reticulum struc- and King-Denborough Syndrome] are thought to arise due
tures, myonuclei were positioned normally (Durieux to ‘leaky’ RyR1 channel activity, channel instability or
et al., 2010). Homozygous mice displayed neonatal lethal- reduced Ca2+ conductance (Wilmshurst et al., 2010).
ity. Cowling et al. (2011) generated mouse models that Recessive mutations may cause misolocalized RyR1 chan-
overexpressed either wild-type or p.Arg465Trp DNM2 nels, or reduced RYR1 abundance (Zhou et al., 2013). KI
using intramuscular delivery of viral vectors. This study RYR1 p.Tyr522Ser (this mutation is responsible for MHS
showed that muscle-specific expression of mutant DNM2 in patients) mice exhibit malignant hyperthermia suscepti-
resulted in a substantial loss of muscle mass, increased in- bility, (MHS)-like responses to halothane and heat stress.
ternal and central nuclei, reduced muscle strength and triad Histologically the muscle is characterized by the step-wise
defects (including abnormal T tubule morphology and development of contracted sarcomeres and core-like re-
swollen sarcoplasmic reticulum). Swollen sarcoplasmic re- gions, secondary to mitochondrial dislocation and triad dis-
ticulum were also observed in myofibres overexpressing ruption (Boncompagni et al., 2009) (structured cores),
wild-type DNM2. followed by sarcomeric disintegration in the core (unstruc-
Cowling et al. (2014) observed that XLMTM patient tured cores). KI p.Tyr522Ser myofibres also display disor-
muscle and Mtm1 / mouse muscle contained elevated ganized T tubules and triad positioning (Boncompagni
levels of DNM2. Subsequently Mtm1 / mice that were et al., 2009). Another KI mouse model (KI p.Ile4898Thr)
hemizygous for Dnm2 were generated. Reduction of of an excitation-contraction coupling RYR1 mutation (a
DNM2 levels, restored the lifespan of Mtm1 / mice, im- common central core disease mutation in patients) exhibits
proved whole-body strength and diaphragm function slowly progressive myopathy with skeletal muscle weak-
(Cowling et al., 2014). Structural abnormalities including ness, impaired mobility and hindlimb paralysis. The mice
myofibre atrophy, nuclear positioning and sarcomere and also developed age-dependent structural lesions within their
triad disorganization, were also reduced in Mtm1 / muscle, sarcomeric misalignment and Z-disc streaming pro-
muscle when DNM2 levels were reduced. This study sug- gressing to mini-cores, cores and then also nemaline bodies
gests that MTM1 and DNM2 regulate skeletal muscle or- (Zvaritch et al., 2009). Thus, this mouse model also
ganization and function in a finely balanced system in models the development of a ‘mixed’ congenital myopathy,
which MTM1 seems to be negative regulator of DNM2 core-rod disease.
252 | BRAIN 2015: 138; 246–268 G. Ravenscroft et al.

The pathogenesis of recessive RYR1 mutations has only been found to underlie a form of tubular aggregate myop-
begun to be elucidated. Zhou et al. (2013) showed that athy (which although being associated with characteristic
RYR1 protein levels are reduced in autosomal recessive morphological findings in the biopsy is not strictly speaking
RYR1 patient muscle, as are DHPR levels, and that the a congenital myopathy) (Bohm et al., 2013a). Given the
localization of the alpha-subunit of the DHPR is also per- crucial roles of the triad and sarcoplasmic reticulum in
turbed in patient muscle and in cultured myotubes targeted muscle function and this apparently widespread perturb-
with siRNA to RYR1 (Zhou et al., 2013). These alter- ation of these entities in the congenital myopathies, there
ations resulted in impaired excitation-contraction coupling is an emerging view that impaired excitation-contraction
and Ca2+ handling. Unexpectedly, IP3 receptors (a down- coupling and Ca2+ homeostasis are central to disease
stream effector of PI3,5P2 which resides in the ER/SR and pathogenesis and may thus represent an important thera-
upon IP3 binding mediates Ca2+ release) were more abun- peutic target across a number of the congenital myopathies.
dant in patient muscle and cultured myotubes deficient for
RyR1, likely representing a secondary upregulation of the Neuromuscular junction defects
IP3-Ca2+ axis. In cultured muscle cells, IP3 receptors are
expressed at the nuclear envelope and give rise to slow The neuromuscular junction (NMJ) is a crucial, deeply in-
nucleoplasmic Ca2+ transients that modulate gene expres- folded, specialization of the muscle membrane. Aberrant
sion (reviewed in Carrasco et al., 2004). While the IP3- NMJs and clinical overlap with the congenital myasthenic
Ca2+ signalling pathway does not play a substantial role syndromes were reported already in the earlier literature, as
in skeletal muscle excitation-contraction coupling under a feature of then genetically uncharacterized CNMs (Sher
normal conditions, it’s upregulation due to RyR1 deficiency et al., 1967; Elder et al., 1983; Fidzianska and Goebel,
and potential effects on gene expression, may contribute to 1994). In light of this and the previous discussion of the
the underlying pathophysiology of this condition and could role of membrane remodelling and shaping, the NMJ has
represent a potential therapeutic target (Zhou et al., 2013). recently come into focus in our understanding of the patho-
genesis of the congenital myopathies, in particular for the
Excitation-contraction coupling and the sarcoplas- CNMs.
mic reticulum in other congenital myopathies
Defects in sarcoplasmic reticulum function have also been Myotubularin
implicated in nebulin (NEB, MIM 161650) deficiency, usu- Dowling and colleagues (2012b) re-examined two mouse
ally associated with the histological phenotype of nemaline models of MTM1-associated disease (Mtm1 KO and KI
myopathy. The sarcoplasmic reticulum Ca2+-ATPase pump p.Arg69Cys) and found that both models showed features
(SERCA) inhibitor, sarcolipin, is upregulated 70-fold in consistent with a defect in neuromuscular transmission.
muscle from Neb KO mice (Witt et al., 2006). Speed of Mtm1 KI mice displayed reduced treadmill-running endur-
Ca2+ uptake by SERCA was significantly slower in sarco- ance and increased grip fatigue compared to wild-type
plasmic reticulum vesicles isolated from Neb KO myofibres mice, indicating exercise intolerance and fatigable muscle
compared to wild-type, contributing to a slower speed of weakness: two clinical indicators of defective neuromuscu-
muscle relaxation (Ottenheijm et al., 2008). Thus it ap- lar transmission. Grip fatigue was attenuated following
pears that nebulin may also be involved in modulating administration of neostigmine [an acetylcholine esterase
sarcoplasmic reticulum Ca2+ handling. (AChE) inhibitor], supporting the presence of a NMJ
Most recently, mutation of the SH3 and cysteine-rich defect. Evaluation of Mtm1 KO and KI p.Arg69Cys
domain 3 gene (STAC3; MIM 615521), encoding a novel muscle highlighted an increase in the area of individual
excitation-contraction coupling protein (Nelson et al., NMJs and a reduction in the complexity of the postsynap-
2013; Reinholt et al., 2013), has been found to underlie tic junctional folds (Dowling et al., 2012b). The authors
Native American myopathy (Horstick et al., 2013). Native showed that trafficking of AChR from the endosomal com-
American myopathy (MIM 255995) is characterized by partment to the sarcolemma was impaired in Mtm1 KO
congenital muscle weakness, susceptibility to malignant myotubes and propose that aberrant regulation of junc-
hyperthermia, joint contractures, dysmorphic features tional membrane turnover and abnormal NMJ trafficking
including ptosis, and non-specific myopathic changes on may underlie the altered ultrastructural appearance and
muscle biopsy (no rods, cores or internally-placed nuclei consequently abnormal function of the NMJs (Dowling
were observed) (Stamm et al., 2008a, b). Thus both pri- et al., 2012b). Treatment of Mtm1 KI p.Arg69Cys mice
mary defects of excitation-contraction coupling (due to with pyridostigmine (another AChE inhibitor) also resulted
RYR1 and STAC3 mutations) and secondary defects of in an improvement in motor function (Dowling et al.,
excitation-contraction coupling (arising from the structural 2012b).
triad abnormalities seen in the CNMs due to mutations in Similarly, in mtm1 morphant zebrafish (Dowling et al.,
DNM2 and MTM1, but also NEB mutations) can be iden- 2009), disorganized NMJs were noted (Robb et al., 2011)
tified in the congenital myopathies. In this context, as noted and movement and provoked swimming behaviour were
above, it is also of interest that dominantly acting muta- dramatically improved upon treatment with an AChE
tions in the sarcoplasmic Ca2+ sensor STIM1 have now inhibitor (edrophonium) (Robb et al., 2011).
Pathophysiology of congenital myopathies BRAIN 2015: 138; 246–268 | 253

Dynamin 2 myopathy to include a myasthenic-like phenotype.


In a zebrafish model overexpressing the p.Ser619Leu muta- Pyridostigmine treatment resulted in improved facial
tion of DNM2, a human mutation associated with severe expression and stamina and decreased ptosis. Again, a
CNM presenting in the neonatal period, severe motor de- detailed analysis of neuromuscular transmission was not
fects were observed (Gibbs et al., 2013). Disorganized possible in these patients leaving the final link open.
NMJs, including abnormal clustering of acetylcholine re- All in all, it seems to be worthwhile to investigate the
ceptors and a reduced number of endplates per myofibre, NMJ, histologically, ultrastructurally, and physiologically,
were a striking feature of this model. Treatment with edro- in models of other congenital myopathies, to assess whether
phonium improved motor function (escape response) of there might be a role for aberrant NMJ function beyond
p.Ser619Leu larvae, but not uninjected controls or wild- the CNMs, and to what extent modulation of neuromus-
type larvae, indicating that AChE inhibition improved the cular transmission may also represent a therapeutic option
phenotype caused by the presence of the mutant dynamin 2 for these patients.
(Gibbs et al., 2013).
Studies of patient biopsies showed abnormal endplate
morphology (irregular and less complex post-junctional
folds) (Fidzianska and Goebel, 1994) and abnormal Mitochondrial abnormalities
AChE staining (Elder et al., 1983). Later, examination of
Abnormal distributions of mitochondria are a well-
five patients harbouring either the DNM2 p.Ser619Leu or
recognized hallmark feature of the congenital myopathies
p.Glu368Lys substitutions, found that two of the patients
that are characterized by the presence of cores or core-like
had electrophysiological recordings suggestive of impaired
regions within myofibres. In fact, cores are defined by being
neuromuscular transmission (Gibbs et al., 2013).
devoid of mitochondria, recognizable by the appropriate
Importantly, Gibbs et al. (2013) and others have reported
stains. Abnormal accumulations, localization and/or ultra-
anecdotally that treatment of MTM1, DNM2 and genetic-
structure of mitochondria have been reported in central
ally-unresolved CNM patients with pyridostigmine, re-
core disease, multi-minicore disease, centronuclear myop-
sulted in improved muscle strength and function, and
athy, and nemaline myopathy (Sanoudou et al., 2006;
reduced fatigability (Liewluck et al., 2011; Robb et al.,
Hnia et al., 2011; Dowling et al., 2012a; Mokbel et al.,
2011). Thus, modulation of neuromuscular transmission
2013) with the possibility that this abnormal localiza-
through AChE inhibition or enhanced acetylcholine release
tion will also lead to abnormal function regionally in
from the presynaptic nerve terminal (as proposed by
muscle.
Dowling et al., 2012b) may represent a viable therapeutic
approach to attenuate the disease severity in patients with
CNMs, that may lead to an improvement in quality of life.
This approach will now need to be evaluated in a con- Ryanodine receptor
trolled clinical trial in patients with CNM. A recessive ryr1 zebrafish model (due to a spontaneous
mutation in the fast-muscle specific ryanodine receptor
The neuromuscular junction in other congenital gene, ryr1b, that results in a premature stop codon)
myopathies showed reduced levels of ryr1 (5–10% of wild-type), de-
NMJ abnormalities may also occur in other congenital fective excitation-contraction coupling and a severe motor
myopathies. Munot et al. (2010) presented two unrelated phenotype (Hirata et al., 2007). The oxidative stress path-
cases of congenital fibre-type disproportion, due to two way was found to be impaired in this model and it was
different heterozygous slow a-tropomyosin (TPM3, MIM confirmed that ryr1 zebrafish had elevated basal oxidant
191030) mutations, which mimicked myasthenic syn- activity arising from mitochondrial-derived reactive
dromes. Single fibre EMG recordings for both cases oxygen species, which were notably not present in dys-
showed a jitter consistent with impaired neuromuscular trophic zebrafish models (Dowling et al., 2012a).
transmission, however detailed histological assessment of Furthermore, cultured RYR1 patient-derived myotubes
the NMJ was not performed. Pyridostigmine administration also displayed features consistent with increased oxidative
resulted in a modest functional improvement in the only stress (Dowling et al., 2012a). The same authors showed
patient that undertook treatment (Munot et al., 2010). that treatment of RYR1 patient myotubes and ryr1 zebra-
Thus, the link between TPM3 mutations and confirmed fish with the antioxidant N-acetylcysteine (NAC) corrected
abnormalities on neuromuscular transmission has to the oxidative stress and restored zebrafish motor function
remain tentative for now. and muscle pathology. These data implicate mitochond-
Most recently, it was shown that pyridostigmine admin- rially-mediated oxidative stress as a contributing factor in
istration resulted in clinical improvement in two siblings the pathophysiology of RYR1-related myopathies and pro-
with recessive RYR1-related congenital myopathy asso- vide important preclinical evidence in support of antioxi-
ciated with striking fatigable ptosis (Illingworth et al., dant therapy as a potential treatment intervention in these
2014). This study extended the phenotype of RYR1-related diseases.
254 | BRAIN 2015: 138; 246–268 G. Ravenscroft et al.

Selenoprotein N1 with abnormal structures and elevated levels of lipofusin


indicative of oxidative stress, in muscle from nemaline my-
Autosomal recessive mutations in selenoprotein N1 opathy mice harbouring a p.Met9Arg mutation in TPM3.
(SEPN1, MIM 606210) underlie some cases of multi- Most recently, subsarcolemmal mitochondrial accumulations
minicore disease and congenital fibre-type disproportion; and a greater variation in the abundance of respiratory chain
in addition to congenital muscular dystrophy with rigid complexes and porin proteins has been described in muscle
spine (MIM 602771) and myopathy with Mallory body- biopsies from some patients harbouring the p.Lys7del muta-
like inclusions (Scoto et al., 2011). SEPN1 is an endoplas- tion in TPM2 compared with healthy individuals (Davidson
mic reticulum glycoprotein that seems more abundant in et al., 2013; Mokbel et al., 2013), indicating a contribution
foetal than adult tissues (Petit et al., 2003). SEPN1 is of mitochondrial dysregulation and cellular stress to the
thought to be involved in Ca2+ homeostasis and protection pathogenesis of some TPM2-related myopathies.
from oxidative stress (Arbogast and Ferreiro, 2010; Castets
et al., 2012).
Sepn1 / mice show normal growth and lifespan and Myotubularin
only minor defects of muscle morphology and function Abnormal mitochondrial localization has also been re-
under basal conditions. However, under physical challenge ported in MTM1 patient cells (Buj-Bello et al., 2008) and
(e.g. forced swimming) Sepn1 / mice developed an obvi- the mtm1 morphant zebrafish (Dowling et al., 2009). In-
ous phenotype characterized by reduced mobility, body ri- depth examination of MTM1 patient muscle and Mtm1
gidity and curvature of the spine - thus recapitulating deficient mouse muscle and cells, found that there was a
aspects of the human disease (Rederstorff et al., 2011). collapse of the mitochondrial network, with the presence of
Despite this phenotype, no cores or core-like regions were enlarged mitochondria containing fewer cristae (Hnia
observed in Sepn1 / muscle, and mitochondria were et al., 2011). Mtm1-deficient cells also had reduced cyto-
ultrastructurally normal (Rederstorff et al., 2011). chrome c oxidase activity and lower ATP levels than
Similar to RYR1 mutant myotubes, cultured SEPN1- wild-type cells (Hnia et al., 2011), suggesting altered mito-
deficient patient myotubes also have a higher basal oxidative chondrial function as a contributing factor to the patho-
activity and patient-derived fibroblasts are more susceptible physiology of MTM1-related CNM. Hnia et al. (2011)
to H2O2 (Arbogast et al., 2009). This elevated sensitivity to identified that in skeletal muscle, the intermediate filament
H2O2 could be mitigated by pretreatment with NAC protein desmin regulates mitochondrial dynamics and
(Arbogast et al., 2009). It has been suggested therefore that morphology through direct interaction with MTM1.
selenoprotein SEPN1 may be involved in buffering reactive These studies found that Mtm1 KO mouse muscle, Mtm1
oxygen species and/or repairing damaged proteins (Castets siRNA-treated C2C12 and MTM1 patient myoblasts, con-
et al., 2012). tained elevated levels of the intermediate filament protein
RYR1 activity is regulated by its oxidative status (Oba desmin, and abnormal desmin aggregates. MTM1 muta-
et al., 2002) and both RYR1 and SEPN1 are enriched in tions that cause CNM and are located within the critical
sarcoplasmic reticulum terminal cisternae fractions (Jurynec region for desmin binding abolished the MTM1-desmin
et al., 2008), which resulted in the hypothesis that SEPN1 interaction. These data suggest that in the absence of
might modulate RYR1 activity. In support of such a con- MTM1, desmin aggregates impair intermediate filament
nection, Ca2+ handling was altered in the sepn1 morphant function and contribute to aberrant internal organelle pos-
zebrafish (Jurynec et al., 2008) and patient myotubes itioning and functioning (including mitochondria and
(Arbogast et al., 2009). nuclei). In this context it is of note that deletion of
However, it remains to be fully explained how SEPN1 desmin in mice also results in altered distribution, morph-
might modulate RYR1 given that SEPN1 is barely detect- ology and activity of mitochondria (Milner et al., 2000).
able in adult myofibres (Castets et al., 2012). Nonetheless, Similar abnormalities are seen in patients with DES-related
there is emerging evidence that links SEPN1 to RYR1 regu- myofibrillar myopathy (MIM 601419) (Reimann et al.,
lation, and both RYR1 and SEPN1 to increased oxidative 2003; Bar et al., 2007).
stress, perhaps via mitochondria. It remains to be seen whether mitochondrial dysfunction
and oxidative stress are more widely applicable to the dis-
Tropomyosins ease process in multiple congenital myopathies and how
much they are able to influence disease severity and
In striated muscle, there are three principal tropomyosin whether they could represent a more widely applicable
isoforms: a-tropomyosinfast (TPM1), a-tropomyosinslow therapeutic target across various disorders.
(TPM3) and b-tropomyosin (TPM2). Tropomyosins inter-
act with the troponin complex to regulate actin-myosin
interactions. Mutations of TPM2 and TPM3 cause congeni-
tal myopathies with nemaline bodies, caps, or fibre-type
Myofibrillar force generation
disproportion (reviewed in Romero and Clarke, 2013). Defects of sarcomeric function and myofibrillar force gen-
Sanoudou et al. (2006) reported enlarged mitochondria eration are the most obvious consequence to consider in
Pathophysiology of congenital myopathies BRAIN 2015: 138; 246–268 | 255

congenital myopathies arising due to defects in genes muscle contraction. It would of course only strengthen
encoding thin filament proteins or proteins interacting fast twitch type fibres, which in congenital myopathies
with or regulating such proteins (Nowak et al., 1999, are frequently fewer in number, but larger (as seen in
2013; Pelin et al., 1999; Laing and Nowak, 2005; fibre-type disproportion, a common histological pattern
Ottenheijm et al., 2012). However, as we will review, the across various congenital myopathies). However, as
exact effects on myofibrillar force generation may differ described later, such treatments could prove to be problem-
from gene to gene and even between different mutations atic in cases where mutations result in increased Ca2+ sen-
in the same gene. sitivity of the myofibrillar apparatus, where such a
treatment could lead to worsening of the condition.
Nebulin
Myofibrillar structure and function has been most exten-
Skeletal muscle alpha-actin
sively studied in patients harbouring mutations in NEB and Over 200 different mutations have been identified in the
the Neb KO (Ottenheijm and Granzier, 2010) and skeletal muscle a-actin gene (ACTA1, MIM 102610), the
NebDExon55 mouse models (Ottenheijm et al., 2013). major component of the thin filament. The majority of
Studies of nebulin-deficient mice (Neb), zebrafish (neb) these disease-associated mutations act in a dominant
and NEB patient muscle, have revealed a greatly impaired manner, causing significant weakness and hypotonia
force-generating capacity (Bang et al., 2006; Witt et al., (Laing et al., 2009), except for a case notably presenting
2006; Lawlor et al., 2011a; Ottenheijm et al., 2012, with muscle stiffness and hypertonia (Jain et al., 2012).
2013; Telfer et al., 2012). Nebulin is thought of as a mo- Rare recessive loss-of-function cases exist and are compat-
lecular ruler determining the length of the actin thin fila- ible with survival because of compensatory upregulation of
ment, thus it is perhaps not surprising that a unifying cardiac a-actin in skeletal muscle (Nowak et al., 2007). A
feature of nebulin-deficient muscle is a greater range in range of structural histological lesions in muscle are asso-
thin filament length, with many sarcomeres containing ciated with ACTA1 mutations, including actin aggregates,
shorter thin filaments, that result in reduced maximal nemaline bodies (sarcoplasmic and intranuclear), caps,
active tension (Bang et al., 2006; Witt et al., 2006; core-like regions, cytoplasmic bodies, dystrophic features,
Ottenheijm et al., 2009, 2013; Telfer et al., 2012). fibre-type disproportion and zebra bodies, while some
Nebulin-deficient myofibrils also have a reduced Ca2+ sen- cases only show minimal changes within the muscle
sitivity of force production (Witt et al., 2006), a slower biopsy (reviewed in Nowak et al., 2013). It remains un-
rate of tension redevelopment and an elevated tension clear how the presence of mutant ACTA1 protein (in dom-
cost, meaning more ATP has to be expended to generate inant cases) resulting in hypotonic or hypertonic
a given tension. This suggests that there is a reduced frac- phenotypes and the absence of ACTA1 (in the recessive
tion of force-generating cross-bridges, which, in turn, con- ‘null’ cases) (Nowak et al., 2007) can give rise to some
tributes to muscle weakness (Ottenheijm et al., 2010; of the same pathologies, such as nemaline bodies. In this
Lawlor et al., 2011a). Recently studies have shown that context it is of note that TPM3-domaint as well as null
in vitro treatment of nebulin-deficient myofibres with a mutations can both give rise to nemaline bodies
fast skeletal muscle troponin activator (CK-2066260) (Lehtokari et al., 2008). This, together with the fact that
enhanced force production at submaximal activating Ca2+ nemaline bodies can also be seen in seemingly unrelated
levels (de Winter et al., 2013; Lee et al., 2013; Ottenheijm genetic entities such as in RYR1 mutations, suggests that
et al., 2013). This compound belongs to a class of fast the development of the nemaline bodies is a result of dys-
skeletal muscle troponin activators that were recently function of the sarcomere rather than due to a direct ef-
shown to augment force development by slowing the rate fect of the presence of mutant proteins or muscle weakness
of Ca2+ dissociation from troponin C (TnC) and enhancing per se.
cross-bridge formation at a given Ca2+ concentration Studies of transgenic mice expressing the nemaline myop-
(Russell et al., 2012). These fast myofibre specific troponin athy-associated ACTA1 mutation p.Asp286Gly have re-
activators may be applicable to a range of skeletal muscle vealed that there is severe muscle weakness resulting in
disorders, as they have also been shown to be effective in early lethality in some mice, mimicking the severity seen
myasthenic patients (Russell et al., 2012). In the context of in patients (Ravenscroft et al., 2011). Studies of isolated
the congenital myopathies this class of compounds might of myofibres and whole muscle found a reduced Ca2+ sensi-
course be particularly beneficial in patients with disorders tivity compared to wild-type (Ravenscroft et al., 2011),
of the thin filament with abnormal contractility such as is likely due to impaired actin–actin interactions within the
the case in some nemaline myopathies. In addition it has filamentous actin (Ochala et al., 2012). A similar decrease
also been suggested that they would be applicable in dis- in the force-frequency relationship (indicative of decreased
orders that manifest defects of excitation-contraction cou- Ca2+ sensitivity) has been found in muscle from another
pling, including patients with core and central nuclear mouse model of nemaline myopathy (Ravenscroft, unpub-
myopathies, where it could be possible to maximize the lished data), the KI Acta1H40Y (p.His40Tyr) line (Nguyen
force produced from the reduced Ca2+ signalling during et al., 2011). In vitro studies of other ACTA1 mutations,
256 | BRAIN 2015: 138; 246–268 G. Ravenscroft et al.

p.Met132Val and p.Phe352Ser, have shown an increase in a compensatory mechanism counteracting some of the
steady-state isometric force production associated with the effect of the diminished force generated by the cross-
substitutions (Marston et al., 2004; Lindqvist et al., 2012). bridges. The sum of this may then underlie the compara-
Drosophila with ACTA1 mutations, in the indirect flight tively mild phenotype of some of these patients, since force
muscles, were flightless and their muscle showed myofibril- generation at submaximal Ca2+ concentrations was com-
lar disorganization (Sevdali et al., 2013). One mutation, parable between patients and healthy controls.
p.Asp292Val, which causes an almost complete absence A similar mechanism to the altered cross-bridge cycling
of Ca2+ activated contraction (Clarke et al., 2007), resides kinetics described for the TPM3 mutations was also
within a region of actin likely to bind tropomyosin and was observed in muscle expressing the p.Arg133Trp substitu-
shown to rescue the hypercontractile phenotype of troponin tion in b-tropomyosin (TPM2) (Ochala et al., 2007).
mutant flies (Sevdali et al., 2013). Jain et al. (2012) re- Marttila et al. (2012) studied nemaline and cap myop-
ported a patient with nemaline myopathy that presented athy-associated mutations in the b-tropomyosin gene
with congenital hypertonia and stiffness, due to a de (TPM2) and showed that some mutations resulted in
novo p.Lys328Asn substitution. It was shown experimen- altered affinity for actin (three showed reduced affinity,
tally, using the in vitro motility assay, that the mutant actin one showed increased affinity) while others showed reduced
extracted from the patient’s muscle biopsy displayed a sub- Ca2+ sensitivity of contractility (p.Glu117Lys and
stantially increased Ca2+ sensitivity (Jain et al., 2012). Thus p.Glu41Lys). Previously, studies of the TPM2 mutant
Ca2+ sensitizers, such as the troponin activators, may be p.Glu41Lys also showed a reduced Ca2+ sensitivity of
applicable to some ACTA1 mutations where there is force production, which could be mitigated by treatment
reduced Ca2+ sensitivity or reduced steady-state force pro- of myofibres with a Ca2+ sensitizer (EMD 57033)
duction, but not others. While the Jain et al. (2012) report (Ochala et al., 2008). A reduced actin binding affinity
is the only published example of a hypertonic actin myop- has also previously been observed with the TPM2
athy, this clinical entity is increasingly recognized and may p.Arg91Gly substitution associated with a distal arthrogry-
also be associated with other nemaline myopathy asso- posis phenotype (Robinson et al., 2007). Most recently,
ciated gene mutations (C.G. Bönnemann et al., unpub- studies of the recurrent TPM2 p.Lys7del mutation asso-
lished data), thus a congenital myopathy should be ciated with a distal arthrogryposis and potentially worsen-
considered even in cases presenting with hypertonia and ing muscle contracture phenotype (Davidson et al., 2013;
stiffness. Mokbel et al., 2013) have shown that myofibres harbour-
ing the mutant b-Tm produce comparable specific forces to
control myofibres but that the Ca2+ sensitivity of contrac-
Tropomyosins tion is greatly increased (Mokbel et al., 2013). Davidson
Given the central role of tropomyosin as the crucial Ca2+- et al. (2013) also showed, by expressing the p.Lys7del mu-
regulated inhibitor of actin-myosin interactions, it was tation in zebrafish, that head-to-tail oligomerization of
thought that tropomyosin mutations would alter sarco- b-tropomyosin polymers was likely to be impaired, that
meric force production and Ca2+ sensitivity. However, the mutant tropomyosin failed to localize properly within
there does not seem to be a simple unifying mechanism the thin filament and that sarcomere length was altered.
associated with these mutations. The most widely studied Actin-b-Tm co-sedimentation assays found that the actin
tropomyosin mutation is the slow a-tropomyosin (a-Tm, binding affinity of the mutant b-Tm was 6-fold lower
TPM3) p.Met9Arg substitution (Laing et al., 1995; than wild-type b-Tm (Mokbel et al., 2013). A study by
Corbett et al., 2001, 2005). Expression of a-Tmp.Met9Arg Marston et al. (2013) showed that all tested TPM2 or
in cardiomyocytes resulted in a greatly reduced Ca2+ sensi- TPM3 mutations residing within repeating structural
tivity compared with cardiomyocytes expressing a-TmWT motifs that bind actin caused a gain-of-function through
(Michele et al., 1999). Analysis of muscle biopsies from increased Ca2+ sensitivity, while mutations outside of
individuals with nemaline myopathy, or congenital fibre- those regions resulted in reduced Ca2+ sensitivity. It will
type disproportion, due to dominant or recessive mutations be important to also correlate the other mutations modelled
in TPM3, revealed that changes in cross-bridge cycling kin- in this study with clinical observations. Interestingly, some
etics underlie the muscular weakness (Ottenheijm et al., TPM2 p.Lys7del patients presented with contractures at
2011). Myofibres from all five TPM3 patients examined birth or developed contractures postnatally (Davidson
in this study, showed a significantly lower rate of force et al., 2013; Mokbel et al., 2013) and one case was
redevelopment and higher tensions costs (as measured by noted to have generalized muscle hypertonia (Mokbel
ATP consumption rates per force production) suggesting et al., 2013), thus increased Ca2+ sensitivity associated
that the proportion of force-generating cross-bridges is di- with specific TPM2 mutations may lead to muscle stiffness
minished in patient myofibres. A striking feature of these with resulting contractures, adding to the differential diag-
TPM3 myofibres was a greatly increased Ca2+ sensitivity, nosis of this phenomenon (Sung et al., 2003; Robinson
resulting in significantly higher pCa50 values (Ottenheijm et al., 2007; Jain et al., 2012; Mokbel et al., 2013). As
et al., 2011). Notably, this increased sensitivity does not alluded to previously, Ca2+ sensitizers (Ochala, 2010) are
lead to muscle stiffness but is rather thought to represent unlikely to be of therapeutic benefit in patients where there
Pathophysiology of congenital myopathies BRAIN 2015: 138; 246–268 | 257

already is enhanced sarcomeric Ca2+ sensitivity and may myocytes and isolated myofibrils harbouring MYH7 muta-
even exacerbate the development of stiffness and contrac- tions were lower than controls, even when normalized to
tures. Thus, knowledge of the exact physiological conse- cross-sectional area. Cardiomyocytes from patients har-
quences of a mutation will be essential in order to select bouring MYH7 mutations were hypocontractile at maximal
potential treatments for a given patient. and submaximal Ca2+ concentrations (Witjas-Paalberends
et al., 2013). Furthermore, cardiomyocytes from MYH7
Myosin patients show increased Ca2+ sensitivity, likely due to hypo-
phosphorylation of PKA and smaller length-dependent ac-
The first skeletal muscle-specific MHC gene associated with tivation (Sequeira et al., 2013); however these features
disease was MYH2. There are a handful of myopathy cases were observed in all cases of hypertrophic cardiomyopathy
in the literature arising due to autosomal dominant or re- (HCM) and are thus likely to be a generalized response to
cessive mutation of the myosin heavy chain IIA gene the cellular remodelling that occurs in hypertrophic cardio-
(MYH2; MIM 160740) (Martinsson et al., 2000; myopathy rather than a direct result of the MYH7 gene
Tajsharghi et al., 2005, 2010; D’Amico et al., 2013). The defect. Similar studies now need to be done for the
phenotype of MYH2 cases can vary greatly with some cases MYH7 mutations associated with skeletal muscle disease.
presenting with multiple joint contractures (Martinsson Autosomal dominant mutations in two genes (MYH3
et al., 2000; de Winter et al., 2013) and severe respiratory and MYH8), encoding developmental MHC isoforms, are
distress at birth (D’Amico et al., 2013) to cases of child- responsible for some presentations of congenital distal
hood or adult onset of mild muscle weakness and myalgia arthrogryposis, which postnatally is not progressive,
(Tajsharghi et al., 2005, 2010). In all cases the disease was thus suggesting a largely developmental mode of action
either non-progressive or was slowly progressive in the (reviewed in Tajsharghi and Oldfors, 2013).
third to fifth decade of life, ophthalmoparesis from birth
was a prominent feature in all cases. In the more rarely
occurring autosomal recessive cases, an absence of MHC
IIA myofibres has been demonstrated (Tajsharghi et al.,
Atrophy
2010). The surprisingly mild skeletal muscle phenotype of Skeletal muscle bulk is determined by the net effect of
the recessive MYH2 null patients is likely due to compen- hypertrophy and atrophy processes: protein synthesis and
sation by the other MHC isoforms and contrasts with the degradation (comprising atrophy and autophagy) (reviewed
profound severity observed in patients with null mutations in Schiaffino and Mammucari, 2011). Atrophy and dis-
of other sarcomeric genes [e.g. ACTA1 (Nowak et al., ordered autophagy are common in a wide variety of
2007), CFL2 (Agrawal et al., 2007; Ockeloen et al., muscle disorders so that it can be challenging to determine
2012) and NEB (Ottenheijm et al., 2009)]. The proportion how specific or non-specific these phenomena are in any
of MHC IIA myofibres increases with age and it is thus given disorder. In the congenital myopathies, gross skeletal
hypothesized that the progression of disease later in life is a muscle atrophy is frequently observed upon clinical exam-
direct result of the increasing proportion of MYH2 mutant ination and, at the level of the myofibre, a common sec-
protein (p.E706K) in the autosomal dominant cases ondary or primary histological abnormality is type I (slow)
(Tajsharghi et al., 2002). Thus, similarly to the autosomal myofibre atrophy (Romero and Clarke, 2013; Wang and
dominant ACTA1 mutations, the mutant protein load Pessin, 2013). An emerging theme, therefore, is the role of
seems to be critical to disease severity. skeletal muscle protein synthesis and degradation molecular
Autosomal dominant mutations in the slow b-myosin pathways in the congenital myopathies; however, it must be
heavy chain gene (MYH7; MIM 160760) are known to cautioned again that there is little direct evidence to sup-
cause two congenital myopathies: hyaline body (myosin port a causative link between the congenital myopathies
storage) myopathy (Bohlega et al., 2004) and multi-mini- and skeletal muscle atrophy.
core disease (Cullup et al., 2012; Clarke et al., 2013), in
addition to Laing distal myopathy (MIM 160500) and car-
diomyopathies. A recent report suggests that MYH7 muta-
Protein degradation pathways
tions may be more frequent in the congenital myopathies There are two main proteolytic systems that control protein
than previously suggested (Clarke et al., 2013). Typically turnover in skeletal muscle: the ubiquitin-proteasome path-
mutations altering amino acid residues within the globular way (atrophy) and the autophagy-lysosome machinery.
head domain cause a cardiomyopathy whereas those alter- Atrophy and autophagy, while distinct programmes, are
ing amino acids within the tail rod domain cause a skeletal both coordinately regulated via the transcription factors
muscle phenotype (Armel and Leinwand, 2009, 2010); FoxO3 and mammalian target of rapamycin (mTOR).
however, in recent years this distinction has become less The ubiquitin-proteasome pathway is responsible for deg-
obvious (Muelas et al., 2010; Homayoun et al., 2011; radation of short-lived soluble proteins and myofibrillar
Lamont et al., 2014). The pathophysiology of MYH7- proteins. Major players in the ubiquitin-proteasome path-
related cardiomyopathies has begun to be elucidated, the way are the E3 ubiquitin ligases (also known as atrogenes),
maximum force generating capacity of single cardiac these include: F-box protein atrogin-1 (also known as
258 | BRAIN 2015: 138; 246–268 G. Ravenscroft et al.

MAFbx, now known as FBXO32) and striated muscle spe- 2010); homozygous mice died within 24 h of birth.
cific tripartite motif (TRIM) proteins, also known as Expression of atrophy- and autophagy-related genes:
MURF (muscle ring finger) proteins (reviewed in Perera FOXO3, MURF1 and Gabarap11 were elevated in 2-
et al., 2012). Of particular interest to the congenital myo- month-old heterozygous KI muscle relative to wild-type.
pathies is the finding that the sarcomeric contractile appar- This study also showed upregulation of FOXO3 and
atus, dysfunction of which is so central to many congenital Gabarap11 in skeletal muscle from a single patient with a
myopathies, is directly involved in regulating atrophy sig- DNM2 mutation (Durieux et al., 2010). Skeletal muscle
nalling. For example, the mechanosensitive M-line titin atrophy was also observed in mouse muscle in which wild-
kinase domain is known to recruit MURF2 to the sarco- type or p.Arg465Trp mutant DNM2 was overexpressed
mere (reviewed in Voelkel and Linke, 2011). Thus it is using adeno-associated virus vectors (Cowling et al., 2011).
conceivable, although not yet proven, that changes in the An interesting example of how a sarcomeric protein can
tension generation on the sarcomere will influence these directly influence muscle protein turnover is supplied by
atrophy pathways. titin and its kinase domain, which regulates a signalling
complex by interacting with Nbr, which regulates
Atrophy in centronuclear myopathy MURF2 via p62/SQSTM1, thus providing a link to trans-
late sensing of mechanical load to regulation of muscle
The role of atrophy in congenital myopathies has been best protein turnover (Lange et al., 2005). A mutation in one
characterized in the centronuclear myopathies. In the Mtm1 recessive titinopathy patient with arthrogryposis, cardiomy-
p.Arg69Cys mouse, skeletal muscle atrophy preceded the opathy and also muscle atrophy has been shown to com-
onset of muscle weakness (Pierson et al., 2012), leading pletely abolish this kinase activity (Chauveau et al., 2014).
the authors to suggest that the atrophy in this model was It will be interesting to see how this pathway will be af-
not a consequence of muscle inactivity but was associated fected in other patients and animal models.
intrinsically with the genetic defect. Joubert and colleagues
(2013) found that intramuscular delivery of an adeno-
associated virus designed to deplete MTM1 resulted in a Atrophy in nemaline myopathy
myotubular myopathy phenotype that included skeletal
muscle atrophy. The pathway by which MTM1 deficiency Mutation of genes encoding BTB-Kelch proteins, known to
leads to atrophy has begun to be elucidated. Elegant studies bind cullin 3 to form E3 ubiquitin ligases (Sambuughin
by Razidlo et al. (2011) showed that in MTM1-depleted et al., 2012; Canning et al., 2013), has been shown to
HeLa cells and myotubes there were decreased levels of cause nemaline myopathies [KBTBD13 (Sambuughin
phosphorylated Akt (activated, Akt-P), and identified that et al., 2010), KLHL40 (Ravenscroft et al., 2013b),
this was mediated by an increase in PI3P levels. Decreased KLHL41 (Gupta et al., 2013)] and an early-onset distal
levels of Akt-P resulted in greater activity of FoxO1 and myopathy (KLHL9) (Cirak et al., 2010); suggesting a
decreased activity of proteins (S6K and 4E-BP1) regulated direct link between impairment of the ubiquitin-proteasome
by mammalian target of rapamycin complex 1 (mTORC1) system and skeletal muscle myopathies. Recently, Garg
of the growth axis (Razidlo et al., 2011). Thus aberrant et al. (2014) showed that Klhl40 KO mice presented with
mTORC1 and FOXO1 signalling may underlie some of a lethal nemaline myopathy-like disease. They also identi-
the pathology of MTM1 deficiency. Indeed mTORC1- fied nebulin and leiomodin 3 (LMOD3) as KLHL40-bind-
deficient mice show skeletal muscle atrophy with numerous ing proteins and showed that loss of KLHL40 resulted in
centrally nucleated myofibres (Bentzinger et al., 2008), reduced abundance of NEB and LMOD3 protein in Klhl40
mimicking some of the hallmark features of MTM1 KO muscle. NEB and LMOD3 were also decreased in
deficiency. muscle biopsies from some KLHL40 patients.
In a Mtmr14 KO mouse model, it was found that age- Intriguingly, co-expression of KLHL40 with either NEB
related muscle wasting, sarcopenia, occurred precociously or LMOD3, resulted in increased abundance of these pro-
(Romero-Suarez et al., 2010). It was also noted that teins, in the absence of a change in transcript levels. The
Mtmr14 expression and MTMR14 protein levels were increase in LMOD3 abundance (in the presence of
decreased in old wild-type muscle, suggesting a role for KLHL40) was similar to that observed when a proteasome
MTMR14 in sarcopenia (Romero-Suarez et al., 2010). inhibitor was added and it was shown that KLHL40
The force deficits and altered Ca2+ handling were similar decreased polyubiquitination of LMOD3 (Garg et al.,
for mature Mtmr14 / (12–14 months of age) and old 2014). Thus despite the widely-held view that BTB-Kelch
wild-type muscle (22–24 months of age), suggesting that proteins mediate protein degradation, it appears that
muscle atrophy in both conditions (Mtmr14 deficiency KLHL40 actually stabilizes the thin-filament proteins NEB
and sarcopenia) may be due to altered Ca2+ signalling and LMOD3, by inhibiting proteasome-mediated degrad-
and feedback from the sarcomere. ation. The absence of NEB in some KLHL40 patient
KI mice, heterozygous for the p.Arg465Trp DNM2 muta- muscle may explain the severity of KLHL40 disease, as
tion, develop skeletal muscle weakness prior to the onset of patients with two null alleles of NEB show a similarly
structural abnormalities and muscle atrophy (Durieux et al., severe phenotype (Wallgren-Pettersson et al., 2002).
Pathophysiology of congenital myopathies BRAIN 2015: 138; 246–268 | 259

These observations of the multifaceted involvement of atrophy (through upregulation of atrogenes and MURF
atrophy have lead to preclinical investigations of treatment proteins) and autophagy.
counteracting atrophy in a number of congenital myopathy
models. In a mouse model of nemaline myopathy (KI
Acta1H40Y), upregulation of two hypertrophic factors Approaches to therapy targeting
[IGF1 and four and a half LIM domains protein 1 hypertrophy/atrophy pathways
(FHL1; Cowling et al., 2008)] or delivery of -tyrosine,
Given the prominence of myostatin in regulating myofibre
improved skeletal muscle strength and pathology (Nguyen
atrophy, modulation of myostatin signalling has been vig-
et al., 2011). Downregulation of components of the ubi-
orously pursued as a potential therapeutic avenue for skel-
quitin-proteasome pathway (based on microarray analysis
etal muscle diseases (particularly the muscular dystrophies;
of transcript levels) were observed in skeletal muscle from
reviewed in Amthor and Hoogaars, 2012); however, some
nemaline myopathy patients suggesting altered protein
studies suggest that myostatin inhibition may be detrimen-
turnover (Sanoudou et al., 2003), the functional signifi-
tal to skeletal muscle function (Amthor et al., 2007;
cance of these alterations remains unclear. There is anec-
Giannesini et al., 2013), perhaps in particular in a muscu-
dotal and some experimental evidence from the TPM3
lar dystrophy in which the muscle is inherently fragile. This
Met9Arg mouse that exercise may be beneficial in nemaline
may suggest that non-dystrophic myopathies with promin-
myopathy (Joya et al., 2004; Wallgren-Pettersson et al.,
ent muscle atrophy may be a better target for myostatin
2011); presumably by minimizing susceptibility to disuse
modulation. Inhibition of the activin receptor IIb in
atrophy (Wallgren-Pettersson, 1989; Wallgren-Pettersson
Mtm1 / mice improved muscle strength and lifespan
et al., 1990; North et al., 1997).
(Lawlor et al., 2011b). Thus modulation of the myostatin
The potential benefits of controlled exercise in individuals
pathway might be of benefit in some congenital myopa-
with congenital myopathies clearly warrant further investi-
thies. It seems that this approach should be considered on
gation. Similarly it remains to be investigated more
a disease-by-disease basis and should take into account all
systematically whether targeted disruption of the ubiqui-
of the different mechanisms resulting in skeletal muscle
tin-proteosomal pathway might represent a potential thera-
atrophy.
peutic target for the congenital myopathies.

Protein synthesis pathways Satellite cells defects


One of the most potent known activators of skeletal hyper- Although satellite cells play a major role during myogenesis
trophy is IGF1 (insulin-like growth factor 1) (Musaro and muscle regeneration, their role in postnatal skeletal
et al., 1999). This hypertrophy requires myofibrillogenesis. muscle maintenance and hypertrophy is controversial. In
Until more recently the mechanism whereby IGF1 mediated a limited number of recent studies a potential role for sat-
myofibrillogenesis remained completely obscure. Takano ellite cells has been implicated in the pathobiology of some
et al. (2010) showed that IGF1 produces its hypertrophic congenital myopathies, but again, the disease specificity of
effects via activation of PI3K-Akt signalling that results in these phenomena will need to be further investigated.
complex formation of N-WASP and nebulin. Nebulin-N- Similar to MTM1 patients, the early stages of myogenesis
WASP complexes were found to mediate actin filament nu- in the Mtm1 KO mouse model seems normal - this has led
cleation from the Z-disc (Takano et al., 2010). Thus IGF1- to the suggestion that there is defective skeletal muscle
induced actin filament formation (via nebulin-N-WASP maintenance or regeneration associated with MTM1 defi-
complexes) is required for muscle maturation and ciency (Buj-Bello et al., 2002). Reduced satellite cell num-
hypertrophy. bers were observed in muscle from Mtm1 KO mice,
The most potent negative regulator of skeletal muscle determined by measuring PAX7 abundance and by flow
hypertrophy is myostatin, a member of the TGF beta-like cytometry to quantify the relative number of myogenic
growth factor family, signalling through activin receptors cells (CD31 /CD45 /CD106 + ) isolated from the muscle.
(Amthor and Hoogaars, 2012). Naturally-occurring and This study also showed that the number of resident satellite
genetically-engineered animal models that are myostatin cells declined with age in the Mtm1 KO muscle. The Mtm1
nulls (Mstn / ) present with a ‘double-muscled’ phenotype. KO progenitor cells showed reduced proliferative capacity
Counterintuitively, despite the plethora of naturally occur- in vitro and in in vivo transplantation experiments and
ing animal models (most notably Belgium Blue and were more susceptible to apoptosis (Lawlor et al., 2012).
Piedmontese cattle, sheep, whippet and mouse) (Stinckens The authors hypothesized that the altered Ca2+ homeostasis
et al., 2011) there is only a single-reported isolated case of due to MTM1 deficiency may cause mitochondrial stress
a MSTN partial loss-of-function patient (Schuelke et al., and thereby activate the mitochondrial-apoptotic pathway.
2004). Myostatin is a negative regulator of Akt and thereby In mouse skeletal muscle depleted of MTM1 postnatally, a
inhibits protein synthesis. In addition to this direct role on 90% reduction in the number of resident satellite cells
protein synthesis, myostatin also signals through SMAD2 was observed (Joubert et al., 2013), these studies suggest
and SMAD3 to activate FOXO3, an inducer of both that exhaustion of the satellite cell population may
260 | BRAIN 2015: 138; 246–268 G. Ravenscroft et al.

contribute to the disease progression associated with profound skeletal muscle atrophy and force deficits
MTM1 deficiency. (Masiero et al., 2009; Masiero and Sandri, 2010).
Exhaustion of the satellite cell population was also a fea- Alterations to autophagy and/or lysosomal function are
ture of the Sepn1 null mouse (Castets et al., 2012); reduced observed in many skeletal muscle diseases, including: colla-
satellite cell numbers have also been observed in SEPN1 gen VI-related muscular dystrophies (in which autophagy
patient muscle and likely contributes to the muscle atrophy seems to underperform), Danon disease, Duchenne muscu-
observed (Castets et al., 2012) suggesting that SEPN1 plays lar dystrophy, Pompe disease and X-linked myopathy with
an essential role in maintenance of these skeletal muscle excessive autophagy (Bonaldo et al., 1998; Grumati et al.,
progenitor cells. 2010; De Palma et al., 2012; Ramachandran et al., 2013)
Recessive mutations in MEGF10 (MIM 612453) were and have also recently been implicated in myofibrillar my-
associated with early onset myopathy, areflexia, respiratory opathy (Kley et al., 2012, 2013). Thus both impaired
distress and dysphagia (EMARDD; MIM 614399) (Logan autophagy and excessive autophagy can result in skeletal
et al., 2011; Boyden et al., 2012). MEGF10 is expressed in muscle disease, highlighting the need for the right balance
quiescent and activated satellite cells and knock-down of between protein synthesis and degradation processes.
Megf10 in mouse muscle resulted in satellite cell depletion Recently, Castets et al. (2013) have shown that sustained
(Holterman et al., 2007). EMARDD patient muscle activation of mTORC1 results in impaired autophagy (via
showed reduced mean myofibre diameter and lacked inhibition of Ulk1, despite FOXO3 activation) and pro-
PAX7 + nuclei (Logan et al., 2011). duced a late onset myopathy in the mouse. Conversely,
Elevated satellite cell numbers were observed in skeletal inhibition of mTORC1, via depletion of Raptor, induced
muscle from patients with nemaline myopathy (Sanoudou autophagy regardless of FOXO inhibition. These novel
et al., 2003) and TPM3 p.Met9Arg transgenic mice data suggest that mTORC1 is, in fact, the major regulator
(Sanoudou et al., 2006), suggesting that satellite cell- of skeletal muscle autophagy (Castets et al., 2013).
activated muscle regeneration might be a feature in some Within the congenital myopathies, autophagy has been
subtypes of nemaline myopathy. The role of satellite cells in most studied in the CNMs. Given that autophagy is essen-
the pathophysiology of nemaline myopathy has not been tially a membrane-driven process and that the primary
investigated further. defect in the CNMs involves membrane remodelling, de-
Myotubes and skeletal myofibres are extremely resistant fective autophagy in the CNMs may be a direct conse-
to apoptosis, and this is thought to be due to expression of quence of the underlying genetic defect.
survival proteins: Akt, PI3K and p21 while satellite cells
express anti-apoptotic Bcl-2 as their primary survival
factor (reviewed in Schwartz, 2008). Recently it was
Centronuclear myopathies
shown that myotubularin regulates Akt-mediated cell sur- A clear link between MTM1 deficiency and autophagy is
vival signalling via elevated levels of PI3P (Razidlo et al., tenuous. PI3P is required for the initiation of autophagy
2011). siRNA silencing of MTM1, in HeLa cells, resulted in and PI3,5P2 is required for its progression. Muscle-specific
a robust activation of caspase-dependent pro-apoptotic sig- deletion of PIK3C3 (the primary kinase responsible for the
nalling that resulted in DNA fragmentation and cell death. production of PI3P) causes a lethal dystrophic phenotype
Knock-down of myotubularin in cultured human myotubes in mice and a severe defect of the autophagolysosomal
also resulted in inhibition of Akt phosphorylation, caspase- pathway (Reifler et al., 2014). Thus turnover of PI3P by
dependent pro-apoptotic signalling and DNA fragmenta- myotubularins may be required for autophagy progression
tion (Razidlo et al., 2011). Thus, there may also be a and it is possible that loss of MTM1 could directly impair
role for aberrantly active apoptosis in the pathogenesis of autophagy. However, a study examining the role of myo-
congenital myopathies. tubularins in autophagy showed that MTM1 was only a
weak activator of the process (Vergne et al., 2009).
Levels of some autophagy-associated transcripts
Autophagy effected by (Gabarap11 and LC3b) are increased in Mtm1 KO
mouse muscle (Fetalvero et al., 2013; Joubert et al.,
membrane targeting and 2013). In Cre-induced Mtm1 depletion in adult skeletal
muscle, levels of LC3-I, LC3-II and p62 proteins were ele-
turnover vated (Joubert et al., 2013), furthermore elevated levels of
The role of autophagy in skeletal muscle homeostasis phosphorylated S6, 4EBP1, Akt and phosphorylated Akt
(reviewed in Sandri, 2010) is less well understood than at- were observed. Similarly, LC3-I, LC3-II, p62 and LAMP2
rophy. Autophagy lysosomes regulate the turnover of long- proteins were more abundant in Mtm1 / muscle than
lived proteins and organelles (Sandri, 2010) and involves wild-type; levels of phosphorylated S6 and Akt were also
BNIP3, Gabarap11, LC3 (now knwon as MAP1LC3A) and elevated in Mtm1 / muscle (Fetalvero et al., 2013).
p62, as well as a host of other players. Autophagy and Elevated levels of LC3, p62, polyubiquitinated proteins
atrophy are inextricably linked; counterintuitively, autop- and dysfunctional mitochondrial are also a feature of
hagy blockade in Atg7 / mouse muscle resulted in Atg7 / null mice (Masiero et al., 2009; Masiero and
Pathophysiology of congenital myopathies BRAIN 2015: 138; 246–268 | 261

Sandri, 2010). This suggests that mTORC1-mediated to the phenotype of KI-Dnm2R465W mice, as opposed to upre-
blockade of normal autophagic degradation may contribute gulated autophagy observed with MTMR14 mutation.
to the impaired autophagy observed in Mtm1 depleted
muscle and partially contributes to the muscle pathology
associated with MTM1 deficiency (Fetalvero et al., 2013; Other possible therapies for
Joubert et al., 2013). However, the autophagic defect in
Mtm1 deficient mouse muscle may be secondary to the congenital myopathies
triad dysfunction, the sarcoplasmic reticulum is a major As we have pointed out throughout the discussion so far,
membrane donor to autophagy, or the general poor understanding the pathomechanisms active in the congeni-
health of these animals. tal myopathies is important in defining targets against
The link between autophagy and MTMR14 is much which to develop therapies, such as compounds that can
clearer, where failed regulation likely of PI3,5P2 results in ameliorate the downstream consequences of the underlying
impaired regulation of autophagic flux. Vergne and col-
genetic defect/s that could modulate diseases. However, tar-
leagues (2009) showed that siRNA knock-down of
geting the primary cause of disease, i.e. the genetic muta-
MTMR14 and loss of its enzymatic activity, in C2C12
tion and their immediate consequences, while challenging,
myoblasts, resulted in the formation of autophagosomes
obviously represents a major goal for arriving at effective
and also facilitated their maturation into autolysosomes.
treatments for these conditions.
LC3-II levels were unchanged following MTMR14 siRNA
treatment in the absence of Atg5 (autophagy-related 5),
showing that MTMR14 negatively regulates autophagy in Enzyme replacement therapy
an Atg5-dependent manner. In support of this, overexpres- Enzyme replacement therapy has been a therapeutic strat-
sing MTMR14 resulted in an increase of p62 aggregates in egy that has been pursued in a number of diseases resulting
transfected C2C12 myoblasts suggesting impaired autop- from enzyme deficiencies (reviewed in Valayannopoulos,
hagy (Vergne et al., 2009). MTMR14 was found to co- 2013). Notably, enzyme replacement therapy is now in
localize with ATG12, ATG16 and LC3-II in early autopha- clinical use for a skeletal muscle disease, glycogen storage
gic organelles and autophagasomes in C2C12 myoblasts. disease II/Pompe disease resulting from deficiency of GAA
Furthermore knock-down of MTMR14 resulted in (Thurberg et al., 2006; Gungor et al., 2013). Lawlor et al.
increased recruitment of the autophagic PI3P-binding pro- (2013) have shown that intramuscular delivery of myotu-
tein WIPI1 (the mammalian homologue of yeast ATG18) bularin-fused with a skeletal muscle penetrating-antibody
(Xie and Klionsky, 2007) to autophagosomes. Finally, protein (3E10Fv-MTM1) resulted in improved muscle con-
Vergne et al. (2009) showed that overexpression of a tractility and pathology and improved gait and ambulation
CNM MTMR14 variant (p.Arg336Gln) within the phos- of Mtm1 KO mice. Recently, AAV8-mediated MTM1 gene
phatase domain did not promote accumulation of p62 therapy was explored in Mtm1 KO mice and dogs carrying
puncta, whereas wild-type and p.Tyr462Cys (a mutant out- a naturally occurring Mtm1 mutation (Childers et al.,
side of the catalytic domain) MTMR14, did increase p62 2014). It was shown that a single intravascular injection
aggregation (Vergne et al., 2009). These data suggest that was sufficient to increased muscle strength, reduce muscle
the CNM p.Arg336Gln substitution renders MTMR14 pathology and extend survival (Childers et al., 2014).
unable to regulate autophagy. These studies demonstrate the efficacy of enzyme replace-
Dowling et al. (2010) found elevated induction of autop- ment therapy and pave the way for clinical trials for myo-
hagy in mtm1/mtmr14 double morphant zebrafish, as tubular myopathy. Similarly, enzyme replacement therapy
determined by increased LC-II levels and vacuolated mem- may also be expected to be of therapeutic benefit in models
branes, with severe effects on gross embryogenesis and of MTMR14-related congenital myopathies although this
extensive autophagic defects. Although accumulation of ab- has not been attempted.
normal membranous structures had previously been re-
ported for mtm1 morphant fish, autophagy was not
altered in these fish (Dowling et al., 2009). Thus, these
Upregulation therapy
authors hypothesize that the phenotype of the double mor- Evidence from patients (Nowak et al., 2007; Ravenscroft
phants results from dysregulated organelle and membrane et al., 2008) and experimental data, concur that mutant
breakdown and overloading of the autophagic system pri- actin load determines disease severity in dominant
marily due to loss of mtmr14 (Dowling et al., 2010). ACTA1 disease (Drummond et al., 1991; Domazetovska
Skeletal muscle and cultured embryonic fibroblasts from et al., 2007; Ravenscroft et al., 2008; 2011, 2013a;
knock-in Dnm2 p.Arg465Trp (KI-Dnm2R465W) mice show Haigh et al., 2010; Sevdali et al., 2013) and thus modula-
elevated levels of LC3, Gabarap11 and p62 transcripts and tion of the amount of mutant ACTA1 represents a target
greater protein abundance (Durieux et al., 2012). The for dominant ACTA1 patients. Furthermore, upregulation
number of p62-positive accumulations and early phagosomes of the foetal isoform of skeletal muscle a-actin (cardiac
were also elevated in KI- Dnm2R465W embryonic fibroblasts. a-actin; ACTC) in patients with recessive loss of ACTA1
These data suggest that impaired autophagy contributes moderates the disease allowing survival of affected children.
262 | BRAIN 2015: 138; 246–268 G. Ravenscroft et al.

Upregulation of ACTC is thus a valid target for upregula- NHMRC Research Fellowship (APP1002147). C.G.B. is
tion or even gene transfer (Nowak et al., 2009; supported by intramural funds of NINDS/NIH.
Ravenscroft et al., 2013a) as it would be less likely to
cause an immune response to the introduced protein as
compared with re-introducing ACTA1.
References
Agrawal PB, Greenleaf RS, Tomczak KK, Lehtokari VL, Wallgren-
Conclusions Pettersson C, Wallefeld W, et al. Nemaline myopathy with minicores
caused by mutation of the CFL2 gene encoding the skeletal muscle
Here we have attempted to highlight emerging patho- actin-binding protein, cofilin-2. Am J Hum Genet 2007; 80: 162–7.
physiological concepts spanning across different genetic Agrawal PB, Pierson CR, Joshi M, Liu X, Ravenscroft G,
and structural entities that traditionally comprise the con- Moghadaszadeh B, et al. SPEG interacts with myotubularin, and
its deficiency causes centronuclear myopathy with dilated cardiomy-
genital myopathies, with the goal of pointing out avenues
opathy. Am J Hum Genet 2014; 95: 218–26.
for therapeutic developments that are based on disease Al-Qusairi L, Weiss N, Toussaint A, Berbey C, Messaddeq N, Kretz C,
mechanisms. It remains to be more precisely determined et al. T-tubule disorganization and defective excitation-contraction
which of the pathophysiological concepts explored herein coupling in muscle fibers lacking myotubularin lipid phosphatase.
are identified in which of the congenital myopathies, and to Proc Natl Acad Sci U S A 2009; 106: 18763–8.
what degree. For instance, in the most well characterized Amburgey K, Bailey A, Hwang JH, Tarnopolsky MA,
Bonnemann CG, Medne L, et al. Genotype-phenotype correlations
models, those for CNMs, there appears to be a role for all in recessive RYR1-related myopathies. Orphanet J Rare Dis 2013; 8:
five of the key themes in the pathobiology of the disease. 117.
It is perhaps necessary, given the wave of animal models Amburgey K, McNamara N, Bennett LR, McCormick ME, Acsadi G,
generated and studied for the congenital myopathies and Dowling JJ. Prevalence of congenital myopathies in a representative
the studies of patient biopsies and cell lines, that standard pediatric united states population. Ann Neurol 2011; 70: 662–5.
Amoasii L, Bertazzi DL, Tronchere H, Hnia K, Chicanne G, Rinaldi B,
operating procedures be developed and implemented for
et al. Phosphatase-dead myotubularin ameliorates x-linked centro-
the systematic characterization of these models and tissues, nuclear myopathy phenotypes in mice. PLoS Genet 2012; 8:
to allow for greater intermodel interpretation and for com- e1002965.
parison of the efficacies of various therapeutic strategies. Amoasii L, Hnia K, Chicanne G, Brech A, Cowling BS, Muller MM,
This has been recognized and implemented for models of et al. Myotubularin and PtdIns3P remodel the sarcoplasmic reticu-
lum in muscle in vivo. J Cell Sci 2013; 126(Pt 8): 10806–19.
muscular dystrophies (Grounds et al., 2008; Nagaraju and
Amthor H, Hoogaars WM. Interference with myostatin/ActRIIB sig-
Willmann, 2009) and spinal muscular atrophy (Willmann naling as a therapeutic strategy for Duchenne muscular dystrophy.
et al., 2011) and is recognized by the presence of standard Curr Gene Ther 2012; 12: 245–59.
operating procedures on the TREAT-NMD webpage Amthor H, Macharia R, Navarrete R, Schuelke M, Brown SC, Otto A,
(http://www.treat-nmd.eu/research/preclinical/). It would et al. Lack of myostatin results in excessive muscle growth but im-
be desirable to expand this approach to models of the con- paired force generation. Proc Natl Acad Sci USA 2007; 104:
1835–40.
genital myopathies.
Arbogast S, Beuvin M, Fraysse B, Zhou H, Muntoni F, Ferreiro A.
Ultimately it seems likely that treatment of the congenital Oxidative stress in SEPN1-related myopathy: from pathophysiology
myopathies will encompass multimodal regimes: e.g. gene to treatment. Ann Neurol 2009; 65: 677–86.
transfer therapy, enzyme replacement therapy, upregulation Arbogast S, Ferreiro A. Selenoproteins and protection against oxida-
of alternative genes, anti-atrophy approaches, in addition tive stress: selenoprotein N as a novel player at the crossroads of
redox signaling and calcium homeostasis. Antioxid Redox Signal
to perhaps AChE inhibitors, and/or Ca2+ sensitizers and/or
2010; 12: 893–904.
anti-oxidants. The considerable overlap of pathobiological Armel TZ, Leinwand LA. Mutations in the beta-myosin rod cause
mechanisms between the various genetic and structural sub- myosin storage myopathy via multiple mechanisms. Proc Natl
groups of the congenital myopathies might indicate that Acad Sci USA 2009; 106: 6291–6.
some of the potential therapies may be effective in a Armel TZ, Leinwand LA. Mutations at the same amino acid in myosin
broad range of the congenital myopathies and beyond. that cause either skeletal or cardiac myopathy have distinct molecu-
lar phenotypes. J Mol Cell Cardiol 2010; 48: 1007–13.
Bang ML, Li X, Littlefield R, Bremner S, Thor A, Knowlton KU, et al.
Nebulin-deficient mice exhibit shorter thin filament lengths and
Acknowledgements reduced contractile function in skeletal muscle. J Cell Biol 2006;
173: 905–16.
We thank our anonymous reviewers for most helpful com- Bar H, Goudeau B, Walde S, Casteras-Simon M, Mucke N,
mentary and advice. Shatunov A, et al. Conspicuous involvement of desmin tail muta-
tions in diverse cardiac and skeletal myopathies. Hum Mutat 2007;
28: 374–86.
Beggs AH, Bohm J, Snead E, Kozlowski M, Maurer M, Minor K,
Funding et al. MTM1 mutation associated with X-linked myotubular myop-
athy in Labrador Retrievers. Proc Natl Acad Sci USA 2010; 107:
G.R. is supported by a National Health and Medical 14697–702.
Research Council of Australia (NHRMC) Early Career Bentzinger CF, Romanino K, Cloetta D, Lin S, Mascarenhas JB,
Researcher Fellowship (APP1035955) and N.G.L. by a Oliveri F, et al. Skeletal muscle-specific ablation of raptor, but not
Pathophysiology of congenital myopathies BRAIN 2015: 138; 246–268 | 263

of rictor, causes metabolic changes and results in muscle dystrophy. Clarke NF. Congenital fiber-type disproportion. Semin Pediatr Neurol
Cell Metab 2008; 8: 411–24. 2011; 18: 264–71.
Bharucha-Goebel DX, Santi M, Medne L, Zukosky K, Dastgir J, Clarke NF, Amburgey K, Teener J, Camelo-Piragua S, Kesari A,
Shieh PB, et al. Severe congenital RYR1-associated myopathy: The Punetha J, et al. A novel mutation expands the genetic and clinical
expanding clinicopathologic and genetic spectrum. Neurology 2013; spectrum of MYH7-related myopathies. Neuromuscul Disord 2013;
80: 1584–9. 23: 432–6.
Bohlega S, Abu-Amero SN, Wakil SM, Carroll P, Al-Amr R, Lach B, Clarke NF, Ilkovski B, Cooper S, Valova VA, Robinson PJ, Nonaka I,
et al. Mutation of the slow myosin heavy chain rod domain under- et al. The pathogenesis of ACTA1-related congenital fiber type dis-
lies hyaline body myopathy. Neurology 2004; 62: 1518–21. proportion. Ann Neurol 2007; 61: 552–61.
Bohm J, Chevessier F, Maues De Paula A, Koch C, Attarian S, Corbett MA, Akkari PA, Domazetovska A, Cooper ST, North KN,
Feger C, et al. Constitutive activation of the calcium sensor Laing NG, et al. An alphaTropomyosin mutation alters dimer pref-
STIM1 causes tubular-aggregate myopathy. Am J Hum Genet erence in nemaline myopathy. Ann Neurol 2005; 57: 42–9.
2013a; 92: 271–8. Corbett MA, Robinson CS, Dunglison GF, Yang N, Joya JE,
Bohm J, Vasli N, Maurer M, Cowling BS, Shelton GD, Kress W, et al. Stewart AW, et al. A mutation in alpha-tropomyosin(slow) affects
Altered splicing of the BIN1 muscle-specific exon in humans and muscle strength, maturation and hypertrophy in a mouse model for
dogs with highly progressive centronuclear myopathy. PLoS Genet nemaline myopathy. Hum Mol Genet 2001; 10: 317–28.
2013b; 9: e1003430. Cowling BS, Chevremont T, Prokic I, Kretz C, Ferry A, Coirault C,
Bonaldo P, Braghetta P, Zanetti M, Piccolo S, Volpin D, Bressan GM. et al. Reducing dynamin 2 expression rescues x-linked centronuclear
Collagen VI deficiency induces early onset myopathy in the mouse: myopathy. J Clin Invest 2014; 124: 1350–63.
an animal model for Bethlem myopathy. Hum Mol Genet 1998; 7: Cowling BS, McGrath MJ, Nguyen MA, Cottle DL, Kee AJ, Brown S,
2135–40. et al. Identification of FHL1 as a regulator of skeletal muscle mass:
Boncompagni S, Rossi AE, Micaroni M, Hamilton SL, Dirksen RT, implications for human myopathy. J Cell Biol 2008; 183: 1033–48.
Franzini-Armstrong C, et al. Characterization and temporal devel- Cowling BS, Toussaint A, Amoasii L, Koebel P, Ferry A, Davignon L,
opment of cores in a mouse model of malignant hyperthermia. Proc et al. Increased expression of wild-type or a centronuclear myopathy
Natl Acad Sci USA 2009; 106: 21996–2001. mutant of dynamin 2 in skeletal muscle of adult mice leads to struc-
Boyden SE, Mahoney LJ, Kawahara G, Myers JA, Mitsuhashi S, tural defects and muscle weakness. Am J Pathol 2011; 178:
Estrella EA, et al. Mutations in the satellite cell gene MEGF10 2224–35.
cause a recessive congenital myopathy with minicores. Cowling BS, Toussaint A, Muller J, Laporte J. Defective membrane
Neurogenetics 2012; 13: 115–24. remodeling in neuromuscular diseases: insights from animal models.
Buj-Bello A, Fougerousse F, Schwab Y, Messaddeq N, Spehner D, PLoS Genet 2012; 8: e1002595.
Pierson CR, et al. AAV-mediated intramuscular delivery of myotu- Cullup T, Lamont PJ, Cirak S, Damian MS, Wallefeld W, Gooding R,
bularin corrects the myotubular myopathy phenotype in targeted et al. Mutations in MYH7 cause Multi-minicore Disease (MmD)
with variable cardiac involvement. Neuromuscul Disord 2012; 22:
murine muscle and suggests a function in plasma membrane homeo-
1096–104.
stasis. Hum Mol Genet 2008; 17: 2132–43.
D’Amico A, Fattori F, Bellacchio E, Catteruccia M, Servidei S,
Buj-Bello A, Laugel V, Messaddeq N, Zahreddine H, Laporte J,
Bertini E. A new de novo missense mutation in MYH2 expands
Pellissier JF, et al. The lipid phosphatase myotubularin is essential
clinical and genetic findings in hereditary myosin myopathies.
for skeletal muscle maintenance but not for myogenesis in mice.
Neuromuscul Disord 2013; 23: 437–40.
Proc Natl Acad Sci USA 2002; 99: 15060–5.
Davidson AE, Siddiqui FM, Lopez MA, Lunt P, Carlson HA,
Canning P, Cooper CD, Krojer T, Murray JW, Pike AC, Chaikuad A,
Moore BE, et al. Novel deletion of lysine 7 expands the clinical,
et al. Structural basis for Cul3 protein assembly with the BTB-Kelch
histopathological and genetic spectrum of TPM2-related myopa-
family of E3 ubiquitin ligases. J Biol Chem 2013; 288: 7803–14.
thies. Brain 2013; 136: 508–21.
Carrasco MA, Jaimovich E, Kemmerling U, Hidalgo C. Signal trans-
De Matteis MA, Luini A. Mendelian disorders of membrane traffick-
duction and gene expression regulated by calcium release from in-
ing. N Engl J Med 2011; 365: 927–38.
ternal stores in excitable cells. Biol Res 2004; 37: 701–12.
De Palma C, Morisi F, Cheli S, Pambianco S, Cappello V, Vezzoli M,
Castets P, Lescure A, Guicheney P, Allamand V. Selenoprotein N in
et al. Autophagy as a new therapeutic target in Duchenne muscular
skeletal muscle: from diseases to function. J Mol Med (Berl) 2012;
dystrophy. Cell Death Dis 2012; 3: e418.
90: 1095–107.
de Winter JM, Buck D, Hidalgo C, Jasper JR, Malik FI, Clarke NF,
Castets P, Lin S, Rion N, Di Fulvio S, Romanino K, Guridi M, et al.
et al. Troponin activator augments muscle force in nemaline myop-
Sustained activation of mTORC1 in skeletal muscle inhibits consti-
athy patients with nebulin mutations. J Med Genet 2013; 50:
tutive and starvation-induced autophagy and causes a severe, late- 383–92.
onset myopathy. Cell Metab 2013; 17: 731–44. Di Paolo G, De Camilli P. Phosphoinositides in cell regulation and
Ceyhan-Birsoy O, Agrawal PB, Hidalgo C, Schmitz-Abe K, membrane dynamics. Nature 2006; 443: 651–7.
DeChene ET, Swanson LC, et al. Recessive truncating titin gene, Domazetovska A, Ilkovski B, Kumar V, Valova VA, Vandebrouck A,
TTN, mutations presenting as centronuclear myopathy. Neurology Hutchinson DO, et al. Intranuclear rod myopathy: molecular patho-
2013; 81: 1205–14. genesis and mechanisms of weakness. Ann Neurol 2007; 62:
Chauveau C, Bonnemann CG, Julien C, Kho AL, Marks H, Talim B, 597–608.
et al. Recessive TTN truncating mutations define novel forms of Dowling JJ, Arbogast S, Hur J, Nelson DD, McEvoy A, Waugh T,
core myopathy with heart disease. Hum Mol Genet 2014; 23: et al. Oxidative stress and successful antioxidant treatment in
980–91. models of RYR1-related myopathy. Brain 2012a; 135: 1115–27.
Childers MK, Joubert R, Poulard K, Moal C, Grange RW, Dowling JJ, Joubert R, Low SE, Durban AN, Messaddeq N, Li X,
Doering JA, et al. Gene therapy prolongs survival and restores func- et al. Myotubular myopathy and the neuromuscular junction: a
tion in murine and canine models of myotubular myopathy. Sci novel therapeutic approach from mouse models. Dis Model Mech
Transl Med 2014; 6: 220ra210. 2012b; 5: 852–9.
Cirak S, von Deimling F, Sachdev S, Errington WJ, Herrmann R, Dowling JJ, Low SE, Busta AS, Feldman EL. Zebrafish MTMR14 is
Bonnemann C, et al. Kelch-like homologue 9 mutation is associated required for excitation-contraction coupling, developmental motor
with an early onset autosomal dominant distal myopathy. Brain function and the regulation of autophagy. Hum Mol Genet 2010;
2010; 133: 2123–35. 19: 2668–81.
264 | BRAIN 2015: 138; 246–268 G. Ravenscroft et al.

Dowling JJ, Vreede AP, Low SE, Gibbs EM, Kuwada JY, Hnia K, Kretz C, Amoasii L, Bohm J, Liu X, Messaddeq N, et al.
Bonnemann CG, et al. Loss of myotubularin function results in T- Primary T-tubule and autophagy defects in the phosphoinositide
tubule disorganization in zebrafish and human myotubular myop- phosphatase Jumpy/MTMR14 knockout mice muscle. Adv Biol
athy. PLoS Genet 2009; 5: e1000372. Regul 2012; 52: 98–107.
Drummond DR, Hennessey ES, Sparrow JC. Characterisation of mis- Hnia K, Tronchere H, Tomczak KK, Amoasii L, Schultz P, Beggs AH,
sense mutations in the Act88F gene of Drosophila melanogaster. et al. Myotubularin controls desmin intermediate filament architec-
Mol Gen Genet 1991; 226: 70–80. ture and mitochondrial dynamics in human and mouse skeletal
Durieux AC, Vassilopoulos S, Laine J, Fraysse B, Brinas L, Prudhon B, muscle. J Clin Invest 2011; 121: 70–85.
et al. A centronuclear myopathy–dynamin 2 mutation impairs Holterman CE, Le Grand F, Kuang S, Seale P, Rudnicki MA. Megf10
autophagy in mice. Traffic 2012; 13: 869–79. regulates the progression of the satellite cell myogenic program.
Durieux AC, Vignaud A, Prudhon B, Viou MT, Beuvin M, J Cell Biol 2007; 179: 911–22.
Vassilopoulos S, et al. A centronuclear myopathy-dynamin 2 muta- Homayoun H, Khavandgar S, Hoover JM, Mohsen AW, Vockley J,
tion impairs skeletal muscle structure and function in mice. Hum Lacomis D, et al. Novel mutation in MYH7 gene associated with
Mol Genet 2010; 19: 4820–36. distal myopathy and cardiomyopathy. Neuromuscul Disord 2011;
Elder GB, Dean D, McComas AJ, Paes B, DeSa D. Infantile centro- 21: 219–22.
nuclear myopathy. Evidence suggesting incomplete innervation. Horstick EJ, Linsley JW, Dowling JJ, Hauser MA, McDonald KK,
J Neurol Sci 1983; 60: 79–88. Ashley-Koch A, et al. Stac3 is a component of the excitation-
Fetalvero KM, Yu Y, Goetschkes M, Liang G, Valdez RA, Gould T, contraction coupling machinery and mutated in Native American
et al. Defective autophagy and mTORC1 signaling in myotubularin myopathy. Nat Commun 2013; 4: 1952.
null mice. Mol Cell Biol 2013; 33: 98–110. Illingworth MA, Main M, Pitt M, Feng L, Sewry CA, Gunny R, et al.
Fidzianska A, Goebel HH. Aberrant arrested in maturation neuromus- RYR1-related congenital myopathy with fatigable weakness, re-
cular junctions in centronuclear myopathy. J Neurol Sci 1994; 124: sponding to pyridostigimine. Neuromuscul Disord 2014; 24:
83–8. 707–12.
Garg A, O’Rourke J, Long C, Doering J, Ravenscroft G, Jain RK, Jayawant S, Squier W, Muntoni F, Sewry CA, Manzur A,
Bezprozvannaya S, et al. KLHL40 deficiency destabilizes thin fila- et al. Nemaline myopathy with stiffness and hypertonia associated
ment proteins and promotes nemaline myopathy. J Clin Invest 2014; with an ACTA1 mutation. Neurology 2012; 78: 1100–3.
124: 3529–39. Joubert R, Vignaud A, Le M, Moal C, Messaddeq N, Buj-Bello A.
Giannesini B, Vilmen C, Amthor H, Bernard M, Bendahan D. Lack of Site-specific Mtm1 mutagenesis by an AAV-Cre vector reveals that
myostatin impairs mechanical performance and ATP cost of contrac- myotubularin is essential in adult muscle. Hum Mol Genet 2013;
tion in exercising mouse gastrocnemius muscle in vivo. Am J Physiol 22: 1856–66.
Endocrinol Metab 2013; 305: E33–40. Joya JE, Kee AJ, Nair-Shalliker V, Ghoddusi M, Nguyen MA,
Gibbs EM, Clarke NF, Rose K, Oates EC, Webster R, Feldman EL, Luther P, et al. Muscle weakness in a mouse model of nemaline
et al. Neuromuscular junction abnormalities in DNM2-related myopathy can be reversed with exercise and reveals a novel myofi-
centronuclear myopathy. J Mol Med (Berl) 2013; 91: 727–37. ber repair mechanism. Hum Mol Genet 2004; 13: 2633–45.
Gibbs EM, Davidson AE, Telfer WR, Feldman EL, Dowling JJ. The Jungbluth H, Sewry CA, Muntoni F. Core myopathies. Semin Pediatr
myopathy-causing mutation DNM2-S619L leads to defective tubu- Neurol 2011; 18: 239–49.
lation in vitro and in developing zebrafish. Dis Model Mech 2014; Jurynec MJ, Xia R, Mackrill JJ, Gunther D, Crawford T,
7: 157–61. Flanigan KM, et al. Selenoprotein N is required for ryanodine re-
Goebel HH, Blaschek A. Protein aggregation in congenital myopathies. ceptor calcium release channel activity in human and zebrafish
Semin Pediatr Neurol 2011; 18: 272–6. muscle. Proc Natl Acad Sci USA 2008; 105: 12485–90.
Grounds MD, Radley HG, Lynch GS, Nagaraju K, De Luca A. Klein A, Lillis S, Munteanu I, Scoto M, Zhou H, Quinlivan R, et al.
Towards developing standard operating procedures for pre-clinical Clinical and genetic findings in a large cohort of patients with rya-
testing in the mdx mouse model of Duchenne muscular dystrophy. nodine receptor 1 gene-associated myopathies. Hum Mutat 2012;
Neurobiol Dis 2008; 31: 1–19. 33: 981–8.
Grumati P, Coletto L, Sabatelli P, Cescon M, Angelin A, Bertaggia E, Kley RA, Serdaroglu-Oflazer P, Leber Y, Odgerel Z, van der Ven PF,
et al. Autophagy is defective in collagen VI muscular dystrophies, et al. Pathophysiology of protein aggregation and extended pheno-
and its reactivation rescues myofiber degeneration. Nat Med 2010; typing in filaminopathy. Brain 2012; 135: 2642–60.
16: 1313–20. Kley RA, van der Ven PF, Olive M, Hohfeld J, Goldfarb LG,
Gungor D, Kruijshaar ME, Plug I, D’Agostino RB Sr, Hagemans ML, Furst DO, et al. Impairment of protein degradation in myofibrillar
van Doorn PA, et al. Impact of enzyme replacement therapy on myopathy caused by FLNC/filamin C mutations. Autophagy 2013;
survival in adults with Pompe disease: results from a prospective 9: 422–3.
international observational study. Orphanet J Rare Dis 2013; 8: 49. Laing NG, Dye DE, Wallgren-Pettersson C, Richard G, Monnier N,
Gupta VA, Ravenscroft G, Shaheen R, Todd EJ, Swanson LC, Lillis S, et al. Mutations and polymorphisms of the skeletal muscle
Shiina M, et al. Identification of KLHL41 mutations implicates alpha-actin gene (ACTA1). Hum Mutat 2009; 30: 1267–77.
BTB-kelch-mediated ubiquitination as an alternate pathway to myo- Laing NG, Nowak KJ. When contractile proteins go bad: the sarco-
fibrillar disruption in nemaline myopathy. Am J Hum Genet 2013; mere and skeletal muscle disease. Bioessays 2005; 27: 809–22.
93: 1108–17. Laing NG, Wilton SD, Akkari PA, Dorosz S, Boundy K, Kneebone C,
Haigh SE, Salvi SS, Sevdali M, Stark M, Goulding D, Clayton JD, et al. A mutation in the alpha tropomyosin gene TPM3 associated
et al. Drosophila indirect flight muscle specific Act88F actin mutants with autosomal dominant nemaline myopathy. Nat Genet 1995; 9:
as a model system for studying congenital myopathies of the human 75–9.
ACTA1 skeletal muscle actin gene. Neuromuscul Disord 2010; 20: Lamont PJ, Wallefeld W, Hilton-Jones D, Udd B, Argov Z, Barboi AC,
363–74. et al. Novel mutations widen the phenotypic spectrum of slow skel-
Hirata H, Watanabe T, Hatakeyama J, Sprague SM, Saint-Amant L, etal/beta-cardiac myosin (MYH7) distal myopathy. Hum Mutat
Nagashima A, et al. Zebrafish relatively relaxed mutants have 2014; 35: 868–79.
a ryanodine receptor defect, show slow swimming and provide Lange S, Xiang F, Yakovenko A, Vihola A, Hackman P, Rostkova E,
a model of multi-minicore disease. Development 2007; 134: et al. The kinase domain of titin controls muscle gene expression
2771–81. and protein turnover. Science 2005; 308: 1599–603.
Pathophysiology of congenital myopathies BRAIN 2015: 138; 246–268 | 265

Lawlor MW, Alexander MS, Viola MG, Meng H, Joubert R, Masiero E, Sandri M. Autophagy inhibition induces atrophy and
Gupta V, et al. Myotubularin-deficient myoblasts display increased myopathy in adult skeletal muscles. Autophagy 2010; 6: 307–9.
apoptosis, delayed proliferation, and poor cell engraftment. Am J Michele DE, Albayya FP, Metzger JM. A nemaline myopathy mutation
Pathol 2012; 181: 961–8. in alpha-tropomyosin causes defective regulation of striated muscle
Lawlor MW, Armstrong D, Viola MG, Widrick JJ, Meng H, force production. J Clin Invest 1999; 104: 1575–81.
Grange RW, et al. Enzyme replacement therapy rescues weakness Milner DJ, Mavroidis M, Weisleder N, Capetanaki Y. Desmin cyto-
and improves muscle pathology in mice with X-linked myotubular skeleton linked to muscle mitochondrial distribution and respiratory
myopathy. Hum Mol Genet 2013; 22: 1525–38. function. J Cell Biol 2000; 150: 1283–98.
Lawlor MW, Ottenheijm CA, Lehtokari VL, Cho K, Pelin K, Mokbel N, Ilkovski B, Kreissl M, Memo M, Jeffries CM, Marttila M,
Wallgren-Pettersson C, et al. Novel mutations in NEB cause abnor- et al. K7del is a common TPM2 gene mutation associated with
mal nebulin expression and markedly impaired muscle force gener- nemaline myopathy and raised myofibre calcium sensitivity. Brain
ation in severe nemaline myopathy. Skelet Muscle 2011a; 1: 23. 2013; 136: 494–507.
Lawlor MW, Read BP, Edelstein R, Yang N, Pierson CR, Stein MJ, Muelas N, Hackman P, Luque H, Garces-Sanchez M, Azorin I,
et al. Inhibition of activin receptor type IIB increases strength and Suominen T, et al. MYH7 gene tail mutation causing myopathic
lifespan in myotubularin-deficient mice. Am J Pathol 2011b; 178: profiles beyond Laing distal myopathy. Neurology 2010; 75:
784–93. 732–41.
Lee E, Marcucci M, Daniell L, Pypaert M, Weisz OA, Ochoa GC, Muhammad E, Reish O, Ohno Y, Scheetz T, Deluca A, Searby C,
et al. Amphiphysin 2 (Bin1) and T-tubule biogenesis in muscle. et al. Congenital myopathy is caused by mutation of HACD1.
Science 2002; 297: 1193–6. Hum Mol Genet 2013; 22: 5229–36.
Lee EJ, De Winter JM, Buck D, Jasper JR, Malik FI, Labeit S, et al. Munot P, Lashley D, Jungbluth H, Feng L, Pitt M, Robb SA, et al.
Fast skeletal muscle troponin activation increases force of mouse fast Congenital fibre type disproportion associated with mutations in the
skeletal muscle and ameliorates weakness due to nebulin-deficiency. tropomyosin 3 (TPM3) gene mimicking congenital myasthenia.
PLoS ONE 2013; 8: e55861. Neuromuscul Disord 2010; 20: 796–800.
Lehtokari VL, Pelin K, Donner K, Voit T, Rudnik-Schoneborn S, Musaro A, McCullagh KJ, Naya FJ, Olson EN, Rosenthal N. IGF-1
Stoetter M, et al. Identification of a founder mutation in TPM3 in induces skeletal myocyte hypertrophy through calcineurin in associ-
nemaline myopathy patients of Turkish origin. Eur J Hum Genet ation with GATA-2 and NF-ATc1. Nature 1999; 400: 581–5.
2008; 16: 1055–61. Nagaraju K, Willmann R. Developing standard procedures for murine
Liewluck T, Shen XM, Milone M, Engel AG. Endplate structure and and canine efficacy studies of DMD therapeutics: report of two
parameters of neuromuscular transmission in sporadic centronuclear expert workshops on “Pre-clinical testing for Duchenne dystrophy”:
myopathy associated with myasthenia. Neuromuscul Disord 2011; Washington, DC, October 27th-28th 2007 and Zurich, June 30th-
21: 387–95. July 1st 2008. Neuromuscul Disord 2009; 19: 502–6.
Lindqvist J, Penisson-Besnier I, Iwamoto H, Li M, Yagi N, Ochala J. A Nance JR, Dowling JJ, Gibbs EM, Bonnemann CG. Congenital myo-
myopathy-related actin mutation increases contractile function. Acta pathies: an update. Curr Neurol Neurosci Rep 2012; 12: 165–74.
Neuropathol 2012; 123: 739–46. Nelson BR, Wu F, Liu Y, Anderson DM, McAnally J, Lin W, et al.
Liu X, Ramjiganesh T, Chen YH, Chung SW, Hall SR, Schissel SL, Skeletal muscle-specific T-tubule protein STAC3 mediates voltage-
et al. Disruption of striated preferentially expressed gene locus leads induced Ca2 + release and contractility. Proc Natl Acad Sci USA
to dilated cardiomyopathy in mice. Circulation 2009; 119: 261–8. 2013; 110: 11881–6.
Logan CV, Lucke B, Pottinger C, Abdelhamed ZA, Parry DA, Nguyen MA, Joya JE, Kee AJ, Domazetovska A, Yang N, Hook JW,
Szymanska K, et al. Mutations in MEGF10, a regulator of satellite et al. Hypertrophy and dietary tyrosine ameliorate the phenotypes of
cell myogenesis, cause early onset myopathy, areflexia, respiratory a mouse model of severe nemaline myopathy. Brain 2011; 134:
distress and dysphagia (EMARDD). Nat Genet 2011; 43: 1189–92. 3513–26.
Maggi L, Scoto M, Cirak S, Robb SA, Klein A, Lillis S, et al. Nicot AS, Laporte J. Endosomal phosphoinositides and human dis-
Congenital myopathies–clinical features and frequency of individual eases. Traffic 2008; 9: 1240–9.
subtypes diagnosed over a 5-year period in the United Kingdom. Nicot AS, Toussaint A, Tosch V, Kretz C, Wallgren-Pettersson C,
Neuromuscul Disord 2013; 23: 195–205. Iwarsson E, et al. Mutations in amphiphysin 2 (BIN1) disrupt inter-
Majczenko K, Davidson AE, Camelo-Piragua S, Agrawal PB, action with dynamin 2 and cause autosomal recessive centronuclear
Manfready RA, Li X, et al. Dominant mutation of CCDC78 in a myopathy. Nat Genet 2007; 39: 1134–9.
unique congenital myopathy with prominent internal nuclei and North KN. Clinical approach to the diagnosis of congenital myopa-
atypical cores. Am J Hum Genet 2012; 91: 365–71. thies. Semin Pediatr Neurol 2011; 18: 216–20.
Marston S, Memo M, Messer A, Papadaki M, Nowak K, North KN, Laing NG, Wallgren-Pettersson C. Nemaline myopathy:
McNamara E, et al. Mutations in repeating structural motifs of current concepts. The ENMC International Consortium and
tropomyosin cause gain of function in skeletal muscle myopathy Nemaline Myopathy. J Med Genet 1997; 34: 705–13.
patients. Hum Mol Genet 2013; 22: 4978–87. Nowak KJ, Ravenscroft G, Jackaman C, Filipovska A, Davies SM,
Marston S, Mirza M, Abdulrazzak H, Sewry C. Functional character- Lim EM, et al. Rescue of skeletal muscle alpha-actin-null mice by
isation of a mutant actin (Met132Val) from a patient with nemaline cardiac (fetal) alpha-actin. J Cell Biol 2009; 185: 903–15.
myopathy. Neuromuscul Disord 2004; 14: 167–74. Nowak KJ, Ravenscroft G, Laing NG. Skeletal muscle alpha-actin
Martinsson T, Oldfors A, Darin N, Berg K, Tajsharghi H, diseases (actinopathies): pathology and mechanisms. Acta
Kyllerman M, et al. Autosomal dominant myopathy: missense mu- Neuropathol 2013; 125: 19–32.
tation (Glu-706 –4Lys) in the myosin heavy chain IIa gene. Proc Nowak KJ, Sewry CA, Navarro C, Squier W, Reina C, Ricoy JR, et al.
Natl Acad Sci USA 2000; 97: 14614–19. Nemaline myopathy caused by absence of alpha-skeletal muscle
Marttila M, Lemola E, Wallefeld W, Memo M, Donner K, Laing NG, actin. Ann Neurol 2007; 61: 175–84.
et al. Abnormal actin binding of aberrant beta-tropomyosins is a Nowak KJ, Wattanasirichaigoon D, Goebel HH, Wilce M, Pelin K,
molecular cause of muscle weakness in TPM2-related nemaline Donner K, et al. Mutations in the skeletal muscle alpha-actin gene in
and cap myopathy. Biochem J 2012; 442: 231–9. patients with actin myopathy and nemaline myopathy. Nat Genet
Masiero E, Agatea L, Mammucari C, Blaauw B, Loro E, Komatsu M, 1999; 23: 208–12.
et al. Autophagy is required to maintain muscle mass. Cell Metab Oba T, Kurono C, Nakajima R, Takaishi T, Ishida K, Fuller GA, et al.
2009; 10: 507–15. H2O2 activates ryanodine receptor but has little effect on recovery
266 | BRAIN 2015: 138; 246–268 G. Ravenscroft et al.

of releasable Ca2 + content after fatigue. J Appl Physiol 2002; 93: Ramachandran N, Munteanu I, Wang P, Ruggieri A, Rilstone JJ,
1999–2008. Israelian N, et al. VMA21 deficiency prevents vacuolar ATPase as-
Ochala J. Ca2 + sensitizers: An emerging class of agents for counter- sembly and causes autophagic vacuolar myopathy. Acta
balancing weakness in skeletal muscle diseases? Neuromuscul Neuropathol 2013; 125: 439–57.
Disord 2010; 20: 98–101. Ravenscroft G, Colley SM, Walker KR, Clement S, Bringans S,
Ochala J, Li M, Ohlsson M, Oldfors A, Larsson L. Defective regula- Lipscombe R, et al. Expression of cardiac alpha-actin spares extrao-
tion of contractile function in muscle fibres carrying an E41K beta- cular muscles in skeletal muscle alpha-actin diseases–quantification
tropomyosin mutation. J Physiol 2008; 586: 2993–3004. of striated alpha-actins by MRM-mass spectrometry. Neuromuscul
Ochala J, Li M, Tajsharghi H, Kimber E, Tulinius M, Oldfors A, et al. Disord 2008; 18: 953–8.
Effects of a R133W beta-tropomyosin mutation on regulation of Ravenscroft G, Jackaman C, Bringans S, Papadimitriou JM,
muscle contraction in single human muscle fibres. J Physiol 2007; Griffiths LM, McNamara E, et al. Mouse models of dominant
581: 1283–92. ACTA1 disease recapitulate human disease and provide insight
Ochala J, Ravenscroft G, Laing NG, Nowak KJ. Nemaline myopathy- into therapies. Brain 2011; 134: 1101–15.
related skeletal muscle alpha-actin (ACTA1) mutation, Asp286Gly, Ravenscroft G, McNamara E, Griffiths LM, Papadimitriou JM,
prevents proper strong myosin binding and triggers muscle weak- Hardeman EC, Bakker AJ, et al. Cardiac alpha-actin over-expres-
ness. PLoS One 2012; 7: e45923. sion therapy in dominant ACTA1 disease. Hum Mol Genet 2013a;
Ockeloen CW, Gilhuis HJ, Pfundt R, Kamsteeg EJ, Agrawal PB, 22: 3987–97.
Beggs AH, et al. Congenital myopathy caused by a novel missense Ravenscroft G, Miyatake S, Lehtokari VL, Todd EJ, Vornanen P,
mutation in the CFL2 gene. Neuromuscul Disord 2012; 22: 632–9. Yau KS, et al. Mutations in KLHL40 are a frequent cause of
Olive M, Goldfarb LG, Lee HS, Odgerel Z, Blokhin A, Gonzalez- severe autosomal-recessive nemaline myopathy. Am J Hum Genet
Mera L, et al. Nemaline myopathy type 6: clinical and myopatho- 2013b; 93: 6–18.
logical features. Muscle Nerve 2010; 42: 901–7. Razidlo GL, Katafiasz D, Taylor GS. Myotubularin regulates Akt-
Ottenheijm CA, Buck D, de Winter JM, Ferrara C, Piroddi N, Tesi C, dependent survival signaling via phosphatidylinositol 3-phosphate.
et al. Deleting exon 55 from the nebulin gene induces severe muscle J Biol Chem 2011; 286: 20005–19.
weakness in a mouse model for nemaline myopathy. Brain 2013; Rederstorff M, Castets P, Arbogast S, Laine J, Vassilopoulos S,
136: 1718–31. Beuvin M, et al. Increased muscle stress-sensitivity induced by
Ottenheijm CA, Fong C, Vangheluwe P, Wuytack F, Babu GJ, selenoprotein N inactivation in mouse: a mammalian model for
Periasamy M, et al. Sarcoplasmic reticulum calcium uptake and SEPN1-related myopathy. PLoS One 2011; 6: e23094.
speed of relaxation are depressed in nebulin-free skeletal muscle. Reifler A, Li X, Archambeau AJ, McDade JR, Sabha N, Michele DE,
FASEB J 2008; 22: 2912–19. et al. Conditional knockout of pik3c3 causes a murine muscular
Ottenheijm CA, Granzier H. New insights into the structural roles of dystrophy. Am J Pathol 2014; 184: 1819–30.
nebulin in skeletal muscle. J Biomed Biotechnol 2010; 2010: Reimann J, Kunz WS, Vielhaber S, Kappes-Horn K, Schroder R.
968139. Mitochondrial dysfunction in myofibrillar myopathy. Neuropathol
Ottenheijm CA, Granzier H, Labeit S. The sarcomeric protein nebulin: Appl Neurobiol 2003; 29: 45–51.
another multifunctional giant in charge of muscle strength optimiza- Reinholt BM, Ge X, Cong X, Gerrard DE, Jiang H. Stac3 is a novel
tion. Front Physiol 2012; 3: 37. regulator of skeletal muscle development in mice. PLoS One 2013;
Ottenheijm CA, Hooijman P, DeChene ET, Stienen GJ, Beggs AH, 8: e62760.
Granzier H. Altered myofilament function depresses force generation Ribeiro I, Yuan L, Tanentzapf G, Dowling JJ, Kiger A.
in patients with nebulin-based nemaline myopathy (NEM2). J Struct Phosphoinositide regulation of integrin trafficking required for
Biol 2010; 170: 334–43. muscle attachment and maintenance. PLoS Genet 2011; 7:
Ottenheijm CA, Lawlor MW, Stienen GJ, Granzier H, Beggs AH. e1001295.
Changes in cross-bridge cycling underlie muscle weakness in patients Robb SA, Sewry CA, Dowling JJ, Feng L, Cullup T, Lillis S, et al.
with tropomyosin 3-based myopathy. Hum Mol Genet 2011; 20: Impaired neuromuscular transmission and response to acetylcholin-
2015–25. esterase inhibitors in centronuclear myopathies. Neuromuscul
Ottenheijm CA, Witt CC, Stienen GJ, Labeit S, Beggs AH, Granzier H. Disord 2011; 21: 379–86.
Thin filament length dysregulation contributes to muscle weakness Robinson P, Lipscomb S, Preston LC, Altin E, Watkins H, Ashley CC,
in nemaline myopathy patients with nebulin deficiency. Hum Mol et al. Mutations in fast skeletal troponin I, troponin T, and beta-
Genet 2009; 18: 2359–69. tropomyosin that cause distal arthrogryposis all increase contractile
Palmio J, Evila A, Chapon F, Tasca G, Xiang F, Bradvik B, et al. function. FASEB J 2007; 21: 896–905.
Hereditary myopathy with early respiratory failure: occurrence in Romero NB, Bitoun M. Centronuclear myopathies. Semin Pediatr
various populations. J Neurol Neurosurg Psychiatry 2014; 85: Neurol 2011; 18: 250–6.
345–53. Romero NB, Clarke NF. Congenital myopathies. Handb Clin Neurol
Pelin K, Hilpela P, Donner K, Sewry C, Akkari PA, Wilton SD, et al. 2013; 113: 1321–36.
Mutations in the nebulin gene associated with autosomal recessive Romero NB, Lehtokari VL, Quijano-Roy S, Monnier N, Claeys KG,
nemaline myopathy. Proc Natl Acad Sci USA 1999; 96: 2305–10. Carlier RY, et al. Core-rod myopathy caused by mutations in the
Perera S, Mankoo B, Gautel M. Developmental regulation of MURF nebulin gene. Neurology 2009; 73: 1159–61.
E3 ubiquitin ligases in skeletal muscle. J Muscle Res Cell Motil Romero-Suarez S, Shen J, Brotto L, Hall T, Mo C, Valdivia HH, et al.
2012; 33: 107–22. Muscle-specific inositide phosphatase (MIP/MTMR14) is reduced
Petit N, Lescure A, Rederstorff M, Krol A, Moghadaszadeh B, with age and its loss accelerates skeletal muscle aging process by
Wewer UM, et al. Selenoprotein N: an endoplasmic reticulum glyco- altering calcium homeostasis. Aging (Albany NY) 2010; 2: 504–13.
protein with an early developmental expression pattern. Hum Mol Royer B, Hnia K, Gavriilidis C, Tronchere H, Tosch V, Laporte J. The
Genet 2003; 12: 1045–53. myotubularin-amphiphysin 2 complex in membrane tubulation and
Pierson CR, Dulin-Smith AN, Durban AN, Marshall ML, Marshall JT, centronuclear myopathies. EMBO Rep 2013; 14: 907–15.
Snyder AD, et al. Modeling the human MTM1 p.R69C mutation Russell AJ, Hartman JJ, Hinken AC, Muci AR, Kawas R, Driscoll L,
in murine Mtm1 results in exon 4 skipping and a less severe et al. Activation of fast skeletal muscle troponin as a potential thera-
myotubular myopathy phenotype. Hum Mol Genet 2012; 21: peutic approach for treating neuromuscular diseases. Nat Med
811–25. 2012; 18: 452–5.
Pathophysiology of congenital myopathies BRAIN 2015: 138; 246–268 | 267

Sambuughin N, Swietnicki W, Techtmann S, Matrosova V, Wallace T, Tajsharghi H, Hilton-Jones D, Raheem O, Saukkonen AM, Oldfors A,
Goldfarb L, et al. KBTBD13 interacts with Cullin 3 to form a func- Udd B. Human disease caused by loss of fast IIa myosin heavy chain
tional ubiquitin ligase. Biochem Biophys Res Commun 2012; 421: due to recessive MYH2 mutations. Brain 2010; 133: 1451–9.
743–9. Tajsharghi H, Oldfors A. Myosinopathies: pathology and mechanisms.
Sambuughin N, Yau KS, Olive M, Duff RM, Bayarsaikhan M, Lu S, Acta Neuropathol 2013; 125: 3–18.
et al. Dominant mutations in KBTBD13, a member of the BTB/ Tajsharghi H, Thornell LE, Darin N, Martinsson T, Kyllerman M,
Kelch family, cause nemaline myopathy with cores. Am J Hum Wahlstrom J, et al. Myosin heavy chain IIa gene mutation E706K
Genet 2010; 87: 842–7. is pathogenic and its expression increases with age. Neurology 2002;
Sandri M. Autophagy in health and disease. 3. Involvement of autop- 58: 780–6.
hagy in muscle atrophy. Am J Physiol Cell Physiol 2010; 298: Takano K, Watanabe-Takano H, Suetsugu S, Kurita S, Tsujita K,
C1291–7. Kimura S, et al. Nebulin and N-WASP cooperate to cause IGF-
Sanoudou D, Corbett MA, Han M, Ghoddusi M, Nguyen MA, 1-induced sarcomeric actin filament formation. Science 2010; 330:
Vlahovich N, et al. Skeletal muscle repair in a mouse model of 1536–40.
nemaline myopathy. Hum Mol Genet 2006; 15: 2603–12. Telfer WR, Nelson DD, Waugh T, Brooks SV, Dowling JJ. Neb: a
Sanoudou D, Haslett JN, Kho AT, Guo S, Gazda HT, Greenberg SA, zebrafish model of nemaline myopathy due to nebulin mutation. Dis
et al. Expression profiling reveals altered satellite cell numbers and Model Mech 2012; 5: 389–96.
glycolytic enzyme transcription in nemaline myopathy muscle. Proc Thurberg BL, Lynch Maloney C, Vaccaro C, Afonso K, Tsai AC,
Natl Acad Sci USA 2003; 100: 4666–71. Bossen E, et al. Characterization of pre- and post-treatment path-
Schara U, Kress W, Bonnemann CG, Breitbach-Faller N, Korenke CG, ology after enzyme replacement therapy for Pompe disease. Lab
Schreiber G, et al. The phenotype and long-term follow-up in 11 Invest 2006; 86: 1208–20.
patients with juvenile selenoprotein N1-related myopathy. Eur J Tosch V, Rohde HM, Tronchere H, Zanoteli E, Monroy N, Kretz C,
Paediatr Neurol 2008; 12: 224–30. et al. A novel PtdIns3P and PtdIns(3,5)P2 phosphatase with an
Schiaffino S, Mammucari C. Regulation of skeletal muscle growth by inactivating variant in centronuclear myopathy. Hum Mol Genet
the IGF1-Akt/PKB pathway: insights from genetic models. Skelet 2006; 15: 3098–106.
Muscle 2011; 1: 4. Toussaint A, Cowling BS, Hnia K, Mohr M, Oldfors A, Schwab Y,
Schuelke M, Wagner KR, Stolz LE, Hubner C, Riebel T, Komen W, et al. Defects in amphiphysin 2 (BIN1) and triads in several
et al. Myostatin mutation associated with gross muscle hypertrophy forms of centronuclear myopathies. Acta Neuropathol 2011; 121:
in a child. N Engl J Med 2004; 350: 2682–88. 253–66.
Schwartz LM. Atrophy and programmed cell death of skeletal muscle. Valayannopoulos V. Enzyme replacement therapy and substrate reduc-
Cell Death Differ 2008; 15: 1163–9. tion therapy in lysosomal storage disorders with neurological ex-
Scoto M, Cirak S, Mein R, Feng L, Manzur AY, Robb S, et al. pression. Handb Clin Neurol 2013; 113: 1851–7.
SEPN1-related myopathies: clinical course in a large cohort of pa- Velichkova M, Juan J, Kadandale P, Jean S, Ribeiro I, Raman V, et al.
tients. Neurology 2011; 76: 2073–8. Drosophila Mtm and class II PI3K coregulate a PI(3)P pool with
Sequeira V, Wijnker PJ, Nijenkamp LL, Kuster DW, Najafi A, Witjas-
cortical and endolysosomal functions. J Cell Biol 2010; 190:
Paalberends ER, et al. Perturbed length-dependent activation in
407–25.
human hypertrophic cardiomyopathy with missense sarcomeric
Vergne I, Roberts E, Elmaoued RA, Tosch V, Delgado MA, Proikas-
gene mutations. Circ Res 2013; 112: 1491–505.
Cezanne T, Laporte J, et al. Control of autophagy initiation by
Sevdali M, Kumar V, Peckham M, Sparrow J. Human congenital
phosphoinositide 3-phosphatase Jumpy. EMBO J 2009; 28:
myopathy actin mutants cause myopathy and alter Z-disc struc-
2244–58.
ture in Drosophila flight muscle. Neuromuscul Disord 2013; 23:
Voelkel T, Linke WA. Conformation-regulated mechanosensory con-
243–55.
trol via titin domains in cardiac muscle. Pflugers Arch 2011; 462:
Sewry CA, Jimenez-Mallebrera C, Muntoni F. Congenital myopathies.
143–54.
Curr Opin Neurol 2008; 21: 569–75.
Wallgren-Pettersson C. Congenital nemaline myopathy. A clinical
Shen J, Yu WM, Brotto M, Scherman JA, Guo C, Stoddard C, et al.
follow-up of twelve patients. J Neurol Sci 1989; 89: 1–14.
Deficiency of MIP/MTMR14 phosphatase induces a muscle disorder
Wallgren-Pettersson C, Donner K, Sewry C, Bijlsma E, Lammens M,
by disrupting Ca(2 + ) homeostasis. Nat Cell Biol 2009; 11: 769–76.
Bushby K, et al. Mutations in the nebulin gene can cause severe
Sher JH, Rimalovski AB, Athanassiades TJ, Aronson SM. Familial
congenital nemaline myopathy. Neuromuscul Disord 2002; 12:
centronuclear myopathy: a clinical and pathological study.
674–9.
Neurology 1967; 17: 727–42.
Wallgren-Pettersson C, Kivisaari L, Jaaskelainen J, Lamminen A,
Stamm DS, Aylsworth AS, Stajich JM, Kahler SG, Thorne LB,
Holmberg C. Ultrasonography, CT, and MRI of muscles in congeni-
Speer MC, et al. Native American myopathy: congenital myopathy
tal nemaline myopathy. Pediatr Neurol 1990; 6: 20–8.
with cleft palate, skeletal anomalies, and susceptibility to malignant
Wallgren-Pettersson C, Sewry CA, Nowak KJ, Laing NG. Nemaline
hyperthermia. Am J Med Genet A 2008a; 146A: 1832–41.
Stamm DS, Powell CM, Stajich JM, Zismann VL, Stephan DA, myopathies. Semin Pediatr Neurol 2011; 18: 230–8.
Chesnut B, et al. Novel congenital myopathy locus identified in Wang Y, Pessin JE. Mechanisms for fiber-type specificity of skeletal
Native American Indians at 12q13.13-14.1. Neurology 2008b; 71: muscle atrophy. Curr Opin Clin Nutr Metab Care 2013; 16:
1764–9. 243–50.
Stinckens A, Georges M, Buys N. Mutations in the myostatin gene Willmann R, Dubach J, Chen K. Developing standard procedures for
leading to hypermuscularity in mammals: indications for a similar pre-clinical efficacy studies in mouse models of spinal muscular atro-
mechanism in fish? Anim Genet 2011; 42: 229–34. phy: report of the expert workshop “Pre-clinical testing for SMA”,
Sung SS, Brassington AM, Grannatt K, Rutherford A, Whitby FG, Zurich, March 29-30th 2010. Neuromuscul Disord 2011; 21: 74–7.
Krakowiak PA, et al. Mutations in genes encoding fast-twitch con- Wilmshurst JM, Lillis S, Zhou H, Pillay K, Henderson H, Kress W,
tractile proteins cause distal arthrogryposis syndromes. Am J Hum et al. RYR1 mutations are a common cause of congenital myopa-
Genet 2003; 72: 681–90. thies with central nuclei. Ann Neurol 2010; 68: 717–26.
Tajsharghi H, Darin N, Rekabdar E, Kyllerman M, Wahlstrom J, Witjas-Paalberends ER, Piroddi N, Stam K, van Dijk SJ, Oliviera VS,
Martinsson T, et al. Mutations and sequence variation in the Ferrara C, et al. Mutations in MYH7 reduce the force generating
human myosin heavy chain IIa gene (MYH2). Eur J Hum Genet capacity of sarcomeres in human familial hypertrophic cardiomyop-
2005; 13: 617–22. athy. Cardiovasc Res 2013; 99: 432–41.
268 | BRAIN 2015: 138; 246–268 G. Ravenscroft et al.

Witt CC, Burkart C, Labeit D, McNabb M, Wu Y, Granzier H, et al. Zorzato F, Fujii J, Otsu K, Phillips M, Green NM, Lai FA, et al.
Nebulin regulates thin filament length, contractility, and Z-disk Molecular cloning of cDNA encoding human and rabbit forms of
structure in vivo. EMBO J 2006; 25: 3843–55. the Ca2 + release channel (ryanodine receptor) of skeletal muscle
Xie Z, Klionsky DJ. Autophagosome formation: core machinery and sarcoplasmic reticulum. J Biol Chem 1990; 265: 2244–56.
adaptations. Nat Cell Biol 2007; 9: 1102–09. Zvaritch E, Kraeva N, Bombardier E, McCloy RA, Depreux F,
Zhou H, Rokach O, Feng L, Munteanu I, Mamchaoui K, Holmyard D, et al. Ca2 + dysregulation in Ryr1(I4895T/wt) mice
Wilmshurst JM, et al. RYR1 deficiency in congenital myopathies causes congenital myopathy with progressive formation of mini-
disrupts excitation-contraction coupling. Hum Mutat 2013; 34: cores, cores, and nemaline rods. Proc Natl Acad Sci USA 2009;
986–96. 106: 21813–18.

Potrebbero piacerti anche