Open access

Myofibrillar Myopathies and the Z-Disk Associated Proteins

Written By

Avnika Ruparelia, Raquel Vaz and Robert Bryson-Richardson

Submitted: 23 November 2011 Published: 22 August 2012

DOI: 10.5772/50110

From the Edited Volume

Skeletal Muscle - From Myogenesis to Clinical Relations

Edited by Julianna Cseri

Chapter metrics overview

3,655 Chapter Downloads

View Full Metrics

1. Introduction

Myofibrillar myopathies (MFMs) are typically autosomal dominant myopathies with late onset progressive muscle weakness and symptoms initially evident in the distal muscle groups. However, there is a significant variability in the presentation of these diseases, with the age of onset ranging from infantile to late seventies; the involvement of the heart, respiratory muscles, distal or proximal muscle groups; and severity covering the full spectrum from mild muscle weakness to premature lethality. Several myopathies were identified with symptoms within this broad spectrum and the recognition of a common pathology allowed the grouping of these diseases under a single term, MFM [1]. Problems in the classification of these disorders still exist, partially due to the wide spectrum of clinical presentation and the lack of detailed analysis of biopsy samples to identify the defining features of MFM.

The defining features of MFM, identified using histological stains and electron microscopy, are the dissolution of muscle fibres and the formation of protein aggregates. Common pathological features of MFM include presence of amorphous, granular, filamentous or hyaline deposits, interstitial fibrosis, fatty infiltration, centrally located nuclei indicative of regeneration, necrosis and muscle degeneration. Displaced membranous organelles are also evident, either in the cytoplasm or within autophagic vacuoles. Affected areas of the cells are frequently devoid of oxidative enzymatic activity and mitochondria can be abnormally shaped and positioned [2-5]. Characterization of the protein aggregates using immunohistochemistry reveals the presence of a wide range of sarcomeric, extracellular, and ubiquitously expressed proteins including Myotilin, Desmin, αB-Crystallin, Filamin C, BAG3, ZASP, Actin, Titin, Myosin, Xin, Dystrophin, sarcoglycans, Plectin, Delsolin, Ubiquitin, Neural cell adhesion modulator, Gelsolin, Syncoilin, Synemin, TAR DNA-binding protein 43, Heat-shock protein 27, and DNAJB2 [6]. Interestingly, α-Actinin, which is the primary Z-disk crosslinker and is associated with many of the Z-disk proteins mutated in MFM, is not detected in these protein aggregates [6,7].

The Z-disk provides an important structural linkage in the transmission of tension and contractile forces along the muscle fibre and has a role in sensing of muscle activity and signal transduction. In line with the identification of the Z-disk as the primary site affected in these myopathies the identification of MFM causing mutations has revealed a very strong association with the Z-disk, with all of the proteins affected being localised to this structure. Mutations have been identified in the intermediate filament (IF) protein Desmin [8], the chaperone αB-Crystallin [9], the structural protein Myotilin [10], the α-Actinin binding protein ZASP[11], the actin binding protein Filamin C[12], and the co-chaperone BAG3[13]. Based on the Mayo Clinic MFM cohort, 14% of MFMs are due to mutations in ZASP, 13% due to Myotilin mutations, 8% Desmin mutations, 5% αB-Crystallin mutations, 4% BAG3 mutations, and 4% due to mutations in Filamin C, with the genetic basis of more than 50% of MFM cases remaining unknown [14].

Whilst subtle differences in morphology and histochemical staining are found to be associated with certain MFM subtypes [3], they are not reliable in identifying the genetic cause of MFM. Ultrastructural studies on the other hand have been shown to be more informative in identifying the subtype of MFM, although repetition with large sample sizes is required to determine the reliability of ultrastructure studies is directing diagnosis[15].

Mutations in any of the identified MFM genes can also result in other forms of myopathy including dilated cardiomyopathy (DCM), restrictive cardiomyopathy (RCM), distal myopathy, spheroid body myopathy (SBM), and limb-girdle muscular dystrophy (LGMD). Whilst mutations can result in different myopathies, within the cases of MFM there is a remarkable consistency in the phenotype regardless of the gene mutated. This unifying pathological presentation suggests a common mechanism of pathology, although the functions of the MFM proteins and how their mutation results in disease are not fully understood. Determination of the mechanism by which these mutation result in disease will not only be important for the development of therapies for these conditions but will also provide insight into the role of these proteins in the muscle and the many functions of the Z-disk. We present an analysis of the literature surrounding each of these proteins and how their mutations result in disease and discuss the implications for MFM and Z-disk function.

Advertisement

2. Desmin and desminopathies

Desmin, named from the word ‘desmos’ which means ‘link’ is a small, 53KDa, IF protein found in skeletal, smooth [16], and cardiac [17] muscle cells. In mature skeletal muscle, Desmin along with other Desmin-binding molecules such as Plectin, links adjacent myofibrils at the Z-disk and binds them to the sarcolemma at the costameric level [18]. Desmin localisation to the intermediate filament, Z-disk, and costamere provides a cytoskeletal network that links the contractile apparatus to the cell membrane and other structural elements of the cell, which is critical for maintaining the integrity of the cell, ensuring force transmission and providing with a pathway for signalling. In order to form a fully functional IF network Desmin connects with different cell structures from the cell membrane to the nuclear envelope. Therefore Desmin interacts with a range of different muscle, non-muscle, and nuclear proteins. At the Z-disk, it interacts with αB-Crystallin (CRYAB) [19] and Nebulin [20,21]. At the periphery of the Z-disk, costameres, nucleus, and neuromuscular junctions Desmin interacts with Vimentin, Synemin [22], Paranemin [23], Desmulsin [24], Lamin [25], Plectin [26], Nestin [27], spectrins [28], and Ankyrin [29]. Deficiency in Desmin not only results in disturbance to the structure of the sarcomere, but also results in striking changes to the cellular morphology, which may have direct implications for muscle function. Desmin knockout mice show abnormal mitochondrial localization, accompanied by an increase in number and size, a rounded shape and distorted membranes, often showing granules and even mineralised bodies [30].

Structurally, Desmin is made up of three domains; an N-terminal head domain, a highly conserved central α-helical core, and a C-terminal tail domain (Figure 1). The central α-helical core, a region responsible for Desmin assembly into IF, is made up of four consecutive helical segments, 1A, 1B, 2A and 2B, which are linked by short non helical linkers [31,32]. These helical domains are made of tandem repeats of a specific seven amino acid sequence that contains the biochemical properties that allow the proper coiling of the protein. Additionally, the 2B helical domain contains a four amino acid insertion, known as the ‘stutter’, critical for Desmin assembly and conserved between many IF proteins [33,34]. Of the 50 Desmin mutations reported so far that result in severe skeletal and/or cardiac muscle defects the majority affect the coiled domains, five affect the head domain and eleven affect the tail domain (Figure 1). Interestingly, no mutations in domain 2A have been reported to date and more than 50% of reported Desmin mutations are in the 2B domain [35]. Although a correlation between the domain mutated and the clinical features of the patients/carriers has been suggested (reviewed in [35]), when the clinical features are analysed in more detail the only correlation that appears to be maintained is the predominance of skeletal muscle defects in patients with mutations in the 2B domain (Table 1 and Table 2).

Figure 1.

Schematic representation of Desmin domains and mutations.

Mutations are coloured accordingly to the disease classification. Note that 3’UTR is not drawn to scale. Mutations are coloured accordingly to the disease classification. Note that 3’UTR is not drawn to scale.

MutationAge of onset /Initial symptomsClinical and pathological features; other studiesRef
c.5G"/>TS2I?SkelSkel: MW. Pathology: abnormal myofibre size. Other studies:in vitro assembly assays showed assembly into a wider IF network; SW13 and MEF cells form a normal IF network.[36,37]
c.137C"/>TS46F?Skel: MW. Pathological studies: abnormal myofibre size. Other studies:in vitro assembly assays showed IF assembly into wider filaments; SW13 cells form aggregates but MEF cells form a normal IF network.[36,37]
c.137C"/>AS46Y?Skel: MW. Pathology: abnormal myofibre size. Other studies:in vitro assembly assays showed assembly into wider IF filaments; SW13 cells show aggregate formation but MEF cells form a normal IF network.[36,37]
c.430A"/>TK144X37CardCard: DCM; AVB.[38]
c.640-2A"/>C? (exon 3 skipping)26-32CardCard: AVB that required pacemaker insertion.[38]
c.1006G"/>TD336Y37CardCard: DCM; AVB; pacemaker insertion.[38]
c.1315G"/>AE439D51CardCard: atrial fibrillation.[38]
c.1325C"/>TT442I27-35SkelSkel: MW and wasting leading to wheelchair dependence; MA. Card: atrial fibrillation and arrhythmia that required pacemaker insertion; fatal heart failure in some cases. Pathology: fibre splitting; internally located nuclei; Desmin-positive aggregates and vacuoles in myofibres. Other studies:in vitro assembly studies showed normal IF; SW13 and C2C12 cells form normal IF network.[39]
c.1346A"/>CK449Tx14SkelSkel: MW. Pathology: abnormal myofibre size. Other studies:in vitro assembly studies show normal filament formation; SW13 and C2C12 cells form normal IF network.[36,39]
c.1360C"/>TR454W(+ Myotilin mutation)15CardSkel: slowly progressive MW. Card: HCM that required Card transplantation. Pathology: Desmin-positive aggregates within myofibres. Other studies:in vitro assembly studies showed formation of short and irregular filamentous structures and aggregates; SW13 cells show aggregate formation and C2C12 form normal IF.[39,40]
c.1379G"/>TS460I29CardSkel: progressive MW and wasting. Card: AVB that required pacemaker implantation. Pathology: abnormal myofibre size; occasional split and regenerating fibres; vacuoles and Desmin-positive aggregates. Other studies:in vitro assembly studies showed normal filament formation; SW13 cells show aggregate formation but C2C12 form normal IF.[39]
c.1413A"/>CX471Y35CardCard: AVB that required pacemaker implantation.[38]

Table 1.

Description of clinical and pathological features of MFM caused by Desmin mutations.

MutationClassificationRef
c.38C"/>TS13FDRM[37,41-43]
c.46C"/>T*R16CRCM[37,44]
c.322G"/>AE108KDCM[45]
c.338A_339GdelQ113fsX115DRM[46]
c.517_537del*del(R173_179)DRM[47,48]
c.638C"/>T (+α-glucosidase mutation)A213VDRM[49,50]
c.639-1G"/>A + c.735+2A"/>Gdel(D214_E245)DRM[44,51-53]
c.719dupAK240fsX243distal myopathy[54]
c.735G"/>C/TE245Ddistal myopathy[49,53,55]
c.893C"/>TS298LDCM[45]
c.934G"/>AD312NDCM[45]
c.1009G"/>CA337PDCM[49,50,52,56,57]
c.1013T"/>GL338RDRM[50]
c.1024A"/>GN342DDRM[43,49,52]
c.1034T"/>CL345PDRM[8,49,58,59]
c.1049G"/>CR350Pdistal myopathy and CM[60]
c.1048C"/>TR350WDCM[45]
c.1064C"/>GE355PDRM[61]
c.1069G"/>CA357PDRM[49,62]
c.1075_1083deldel(E359_S361)DRM[63]
c.1078G"/>C*A360PDRM[49,52,56]
c.1097_1099deldel(N366)DRM[63,64]
c.1099A"/>TI367FDRM[65]
c.1109T"/>CL370PDRM[49,62,66]
c.1130T"/>CL377PDRM[67]
c.1154T"/>CL385PDRM[49,68]
c.1166A"/>CQ389PDRM[49,69]
c.1175T"/>CL392PDRM[65]
c.1178A"/>T*N393I#DRM[49,50,52,56]
c.1195G"/>TD399YDRM[49,50]
c.1201G"/>AE401KDRM[50]
c.1216C"/>TR406WDRM[44,49,52,65,69,70]
c.1237G"/>AE413KDRM[39,49,71]
c.1255C"/>TP419SDRM[65]
c.1353C"/>GI451MDRM[52,72-74]
c.1358C"/>TT453IDRM[44]
c.1375G"/>AV459IDCM[45]
c.1405G"/>AV469MDCM[37,41-43]

Table 2.

Human myopathies caused by Desmin mutations excluding MFM.

Mutations in Desmin result in many different myopathies (Table 1 and Table 2). Desmin-related myopathies (DRM) is a term that has been used to describe myopathies due to mutations in Desmin and CRYAB including MFMs, here we only use it to refer to those caused by Desmin mutations. In addition to the broad spectrum of DRM, Desmin mutations have also been classified as MFM, distal myopathy, DCM, and RCM (see Table 1 and Table 2). Some of the DRMs may be examples of MFM but without further information it is not possible to re-classify them as MFMs. There is significant cardiac involvement in many Desmin myopathies and in some cases individuals with the same mutation may initially present with cardiac or skeletal muscle symptoms suggesting there is significant phenotypic variability and the possibility of modifiers of the Desmin myopathies. For example, the I451M mutation has been reported in a case of familial DCM without skeletal muscle phenotypes [72] and in individuals with skeletal myopathy without any evident cardiac defect [74]. Furthermore, the mutation was not fully penetrant in the family with DCM [72]. Potential modifiers include -Glucosidase, with a single individual identified as a compound heterozygote for -Glucosidase missense mutations and heterozygous for the Desmin A213V mutation displaying progressive muscle weakness not evident in related individuals carrying A213V alone [50], and Lamin A, as identified in an individual with Emery Dreifuss muscular dystrophy due to heterozygous Lamin A and Desmin V469M mutations [75].

Mutations in Desmin may also affect its capability to interact with its binding partners. Indeed, analysis of mutant protein E245D using solid phase binding assays showed that it binds to Nebulin with increased affinity, reducing Nebulin at the Z-disk, and is more prone to aggregate formation [76]. This interferes with Nebulin’s ability to regulate the thin filament and results in disease [76]. Furthermore, the targeted mutation K190A, not yet observed in disease, shows decreased affinity for Nebulin resulting in decreased targeting of Nebulin to the Z-disk, its accumulation in aggregates in both skeletal and cardiac muscle, and narrower Actin bundles. It was therefore hypothesised that the lack of functional Desmin prevents Nebulin from stabilising Actin thin filaments thereby resulting in collapse of the contractile apparatus [21].

To study the role of Desmin in muscle function two independent knockout mice lines were created [77,78]. Both lines develop normally, are viable and fertile, with no defects in myogenesis. However, they present postnatal multisystem disorder, decreased myofibril alignment, defects in nuclear and mitochondrial positioning within the cell, and severe cardiac degeneration [77-81]. Muscle of Desminknockout mice was also found to be more susceptible to damage following contraction [82]. These studies show that the absence of Desmin does not impair muscle formation or animal viability however, it is important for muscle function and integrity. In addition to the abnormal localisation of mitochondria described in the knockout mouse [81], Desmin mutations, such as K240fsX243, R350P, and E413K, can result in abnormal localisation and function of the mitochondria resulting in a deficiency in oxygen metabolism which impairs muscle function and may contribute to muscle degeneration [49,54,71].

It has been extensively suggested that Desmin may be essential in lateral force transmission by connecting adjacent sarcomeres, and even neighbouring myofibres, by costamere-extracellular matrix (ECM) binding. Therefore, Desmin mutations may impair its ability to respond to applied strain. Studies on Desmin with tail domain mutations in which the filament assembly is normal in both in vitro cell cultures and in transfected cells showed altered flexibility, with significantly increased stiffness compared to wildtype IF. This altered intrinsic properties of IF is hypothesised to prevent Desmin from responding to excess strain thereby resulting in muscle pathology [40]. This is also supported by the del(Arg173-Glu179) knock-in mouse. Detailed analysis of the myocardium of these mice revealed the presence of aggregates containing Desmin and other muscle proteins, characteristic of desminopathies, which disturbed overall IF structure and compromised myocardium function both during baseline conditions and during maximal adrenergic stimulation[83].

The analysis of filament formation in vitro has identified a clear mechanism by which Desmin mutations may disrupt its assembly into filaments.However, it is still not clear which of the many roles of the IF contribute to pathology in desminopathies. The association of Z-disk proteins with MFM may suggest that it is the role of the IF at the Z-disk that is most relevant to these conditions but mitochondrial organisation and tethering of the myofibrils to the sarcolemma have clear links to muscle function and maintenance. The emerging application of whole genome and exome sequencing to mutation detection may improve identification of modifiers of pathology providing an alternative route to examine Desmin function, explain the phenotypic variations observed, and develop areas of potential therapy.

Advertisement

3. B-crystallin and B-crystallinopathies

To date 15 mutations in CRYAB have been reported. CRYAB belongs to the small Heat shock family of proteins (sHSP). It interacts with A-Crystallin (CRYAA) via non-covalent bonds to form large heterogeneous macromolecular complexes [19]. Both CRYAA and CRYAB are found in high levels in the lens tissue of the eye where they are involved in maintaining lens transparency and refractive index [84]. CRYAB is also found in significant amounts in non-lenticular tissues such as skeletal and cardiac muscle, the kidney, and the brain [85-87]. In skeletal muscle CRYAB expression is highest in the oxidative slow twitch muscle and lowest in the glycolytic fast muscle [85,88]. In skeletal and cardiac muscle CRYAB is localised to the Z-disk [89] where it interacts with the I-band protein Actin [19] and various IF proteins including Desmin [19], Vimentin [90], and Glial fibrillary acidic protein(GFAP) [91].

The N-terminal globular domain and the highly conserved C-terminal ‘-crystallin domain’ (ACD; Figure 2) of CRYAB are critical for its chaperone-like function [92-94] and dimerisation [95]. CRYAB prevents stress induced aggregation of various proteins including β- and γ-Crystallins [93,93], Desmin [19], Vimentin [90], and GFAP [91]. Following stressful conditions such as osmotic stress, metal toxicity [96], serum starvation, hypertonic stress, and heat shock [90] CRYAB expression is up-regulated and recruited to the IF to remodel the IF network [90]. Mutations in CRYAB have been shown to interfere with both its dimerisation and chaperone functions. Resolution of the crystal structure of the MFM causing R120G mutant protein showed a disruption to its tertiary structure predicted to interfere with its dimerisation and result in the formation of large soluble oligomers [97]. Moreover, the ACD domain of mutant CRYAB adopts an irregular structure, which decreases its chaperone function, makes it unstable and promotes its aggregation [98-100]. The mutant CRYAB has also been shown to have a higher dissociation constant, which prevents its dissociation from Desmin [101] resulting in Desmin containing aggregates as seen in MFM [100,101]. Therefore, alterationsin the structure of CRYAB, its inability to perform its chaperone functions, and disruptionof its interaction with its binding partners all contribute to disease pathology. In contrast to other MFM genes there does appear to be some correlation between genotype and phenotype with mutations in exon one resulting in isolated cataracts whilst exon three mutations can result in cataracts [102-107], MFM [108-110], distal myopathy [111],and/or CM [112,113], with two mutations resulting in both cataract formation and muscle failure [114,115] (Table 3).

Following contraction CRYAB is phosphorylated and translocates from the cytoplasm to the Z-disk which is thought to allow CRYAB mediated repair or protection of the Z-disk [125]. Recently, a rare case of infantile onset MFM was identified due to a homozygous frameshift mutation, S115fsX14, resulting in muscle stiffness [110]. The authors suggest the mutation results in a loss of contraction timulated translocation to the Z-disk and consequent reduction in muscle repair. CRYAB has also been implicated in indirectly preventing apoptosis and autophagy, inhibiting Caspase 3 mediated [126], Ras induced[127], and Bcl-2 mediated apoptosis [128]. It is therefore not surprising that CRYAB deficiency results in decreased cell viability and an increase in apoptosis in CRYAB knock-out mouse [129] and in patients suffering with CRYAB mutations [108]. In basal breast cancer CRYAB behaves as an oncoprotein [130] and in highly migratory glioma cells prevents apoptosis [131] making it a potential target in cancer therapy. Up-regulation of CRYAB may be part of a general protective mechanism since CRYAB is up-regulated in various pathological conditions such as cardiac ischemia [132], multiple sclerosis [133], Alzheimer’s [134], and other neurodegenerative disorders [84].

Wildtype CRYAB has the capability to bind to mutant protein to prevent its aggregation. For example, transfection of the MFM causing R120G mutant CRYAB into PtK2 cells results in the formation of aggregates but co-transfection with wildtype CRYAB, or the chaperone molecule Hsp27, results in a significant decrease in the amount of insoluble R120G CRYAB present in the cell and the frequency of aggregate formation [135]. Recently the BAG3 co-chaperone protein has also been shown to co-oligomerise with mutant CRYAB to suppress its aggregation and toxicity [136]. This does highlight that stimulating an increase in wildtype CRYAB, Hsp27, or BAG3 may be sufficient to prevent the formation of protein aggregates.

The observation that there is a 10 fold increase in CRYAB expression in differentiating and proliferating myoblasts [137,138] suggests that CRYAB may play a role in regulating myogenesis. CRYAB has been shown to decrease the synthesis, and increase the degradation, of MyoD, a myogenic regulatory factor that specifies cell lineage, resulting in delayed differentiation. Additionally, up-regulation of CRYAB in muscle cells results in sustained expression of cell cyclemarkers such as Cyclin D1 indicating cells were more proliferative [139]. Therefore, CRYAB can influence myogenesis by altering MyoD levels and cell cycle exit. Despite the potential for CRYAB to regulate muscle differentiation CRYAB knockout mice have normal muscle at birth but present with severe muscular dystrophy by week 40 suggesting CRYAB is not critical for muscle development but is essential for muscle function [129].

MutationAge of onset /
Initial symptoms
Clinical and pathological features; other studiesRef.
c.61delA*
S21fcX44
Birth-11 weeks
Resp
Skel: hypertonia. Pathology: presence of necrotic and regenerating fibres; atrophic and splitting myofibres and internally located nuclei; Desmin-, Myotilin-, and some Ubiquitin-positive aggregates at the periphery of myofibres, vacuoles and deposits. Classified as MFM[9,109]
c.325G"/>C
D109H
35-45SkelSkel: MW. Card: DCM; Cataract formation. Pathology: abnormal myofibre size; atrophic and splitting myofibres and internally located nuclei; Desmin-, CRYAB-, and Myotilin-positive aggregates and vacuoles. Classified as MFM[114]
c.343delT*
S115fsX14
4 months
Skel
Skel: muscle stiffness. Pathology: muscle fibrosis. Classified as MFM[110]
c.358A"/>GR120G?SkelSkel: MW; Card: HCM; Cataract formation. Pathology: presence of Desmin and CRYAB aggregates. Other studies: altered CRYAB quaternary structure; Partial unfolding exposes hydrophobic regions thus increases susceptibility to proteolysis and aggregation; disrupted protein binding; HeLa cells show hyperphosphorylation mutant CRYAB and accumulation in the cytoplasm; in vitro studies show Desmin and CRYAB aggregates in the cytoplasm and around the nuclei; rat cardiomyocytes with perinuclear aggregates containing Ubiquitin, β-Tubulin and Hsp25; cardiomyocytes in culture expressing mutant CRYAB show that the contractile apparatus does not work properly; mutant mice myofibrils alignment are impaired, CRYAB and in some cases Desmin- positive aggregates; cardiac hypertrophy; mitochondrial architecture and alignment are altered in cardiomyocytes; mice die by early adulthood. Classified as DRM[98,99,115-122]
c.451C"/>TQ151X43SkelSkel: slowly progressive MW and MA. Pathology: severe abnormal myofibre size; necrotic and regenerating myofibres and internally located nuclei; abnormal Z-disks were detected; Desmin-, CRYAB- and Dystrophin-positive aggregates. Other studies:in vitro assays show that this mutation prevents oligomerisation, without changing its function, but aggregation is enhanced; in vitro assembly assays and COS-7 cells and cardiomyocytes cultures showed an increased tendency to hyperphosphorylation and aggregate formation. Classified as MFM[108,123,124]
c.460G"/>AG154S48-68Skel or CardSkel: slowly progressive MW and MA Card: DCM; moderate VEFR. Pathology: Desmin- and CRYAB-positive aggregates in subsarcolema and in the centre of the myofibres; Z-disk disorganization and smearing, with accumulation of vacuoles and other material. Classified as DCM[111,112]
c.464CTdel
L155fsX163
52
Resp
Skel: MW. Pathology: abnormal myofibre size; fibre degeneration; presence of vacuolations and inclusions; disruption of the intermyofibrillar architecture; Desmin-, Myotilin- and CRYAB-positive aggregates. Classified as MFM[108]
c.470G"/>AR157H40'sCardCard: ventricular tachycardia. Other studies: rat cardiomyocytes show decreased CRYAB binding to Titin in the cardiac specific domain, without affecting its distribution in the cell. Classified as DCM[113]

Table 3.

Description of clinical and pathological features of B-crystallinopathies.

Figure 2.

Schematic representation of CRYAB protein structure and myopathy mutations.

Advertisement

4. Myotilin and myotilinopathies

The first description of myotilinopathy was a missense mutation in a family with LGMD1A [10]. Since this initial discovery, nine additional mutations in Myotilin (myofibrillar protein with Titin-like immunoglobulin domains) have been implicated in LGMD1A [10,140-142], MFM [143], SBM [144] or late onset distal myopathy [145] with all mutations described to date displaying an autosomal dominant pattern of inheritance. One of the mutations identified, S55F, has been found as a cause of both LGMD [141] and MFM [143] suggesting there may be modifiers of the disease that determine the symptoms produced or that there is an overlap in the classification of these conditions that needs to be resolved. Distinctions between these conditions are not clear, since the presence of protein aggregates is associated with MFM and SBM with weakness of distal muscle groups thought to be associated with MFM [143] and proximal muscle groups with LGMD (Table 4).

Myotilin belongs to the immunoglobulin domain containing Actin binding protein family that also contains the Actin organizing proteins Palladin and Myopalladin [146,147]. Myotilin is predominantly expressed in skeletal and cardiac muscle, with the highest levels present in the skeletal muscle. Expression is also detectable at low levels in the peripheral nerves, bone marrow, liver, thyroid gland and lung [153,154]. In skeletal muscle Myotilin is present in both slow type I and fast type II fibers [151] and is localised to the Z-disk [153], although some reports have suggested Myotilin may also be found at the sarcolemma [10,153,155]. A role at the sarcolemma is also supported by the inclusion of Dystrophin in the protein aggregates found in MFM and LGMD1A [10,143]. Like many other Z-disk proteins Myotilin is very dynamic as demonstrated by fluorescent recovery after photobleaching (FRAP) experiments in quail skeletal muscle that showed that 80% of Myotilin in the Z-disk is replaced within five minutes of bleaching [156].

Myotilin contains two identified domains, both essential for its function; a serine rich N-terminal domain, that shares no homology with any known protein, and a C-terminal domain consisting of two Ig-like domains that share high homology to two Z-disk associated Ig-like domains of the giant protein Titin [153] (Figure 3).Seven of the eight identified Myotilin mutations, including the three MFM mutations [143], are in the serine rich domain with one mutation in the second Ig domain [142]. The serine rich domain consists of a stretch of hydrophobic residues that are believed to direct the localisation of Myotilin to the sarcolemma [10]. The serine rich domain is also responsible for the interaction of Myotilin with a range of proteins including the primary Z-disk crosslinker -Actinin [153], Filamin- Actin- and Telethonin-binding protein of the Z-disk (FATZ, Myozenin, Calsarcin) [157], ZASP/Cypher [158], Filamin C [157,159] and the ubiquitin ligases MURF-1 and MURF-2 [160]. Interaction of Myotilin with FATZ directly or indirectly directs the localisation of FATZ to the Z-disk [157]. Myotilin also links Filamin C, found at the periphery of the Z-disk, to -Actinin and anchors the Actin containing thin filaments to the Z-disk thereby providing stability to the sarcomere

MutationAge of onset /Initial symptomsClinical and pathological features; other studiesRef
c.17G"/>AR6H40SkelSkel: progressive MW, culminated in wheelchair dependence. Pathology: abnormal myofibre size and fibrosis; necrotic fibres with macrophage invasion; internally located nuclei; mitochondria aggregation. Classified as LGMD1A[140]
c.116C"/>TS39FChildhood-60sSkelSkel: progressive MW, in some cases wheelchair dependence. Pathology: spheroid bodies with Myotilin immunoreactivity at the periphery. Classified as spheroid body myopathy[144]
c.164C"/>TS55F48-53SkelSkel: slowly progressive to severe MW and wasting. Pathology: abnormal myofibre size with deposits and vacuoles; atrophic and necrotic myofibres; Myotilin-, CRYAB-, Dystrophin-, Desmin-. and Ubiquitin-positive aggregates; clusters of mitochondria. Classified as LGMD1A and MFM[141,143,148-150]
c.170C"/>TT57I27SkelSkel: progressive MW. Pathology: abnormal myofibre size; myofibre degeneration and splitting; centrally located nuclei; vacuoles; Z-disc streaming. Other studies: mice reproduce human MFM pathology: Myotilin-, Desmin-, Ubiquitin-, and Actin-positive aggregates; fibrosis; Z-disk streaming and sarcomere disorganisation; some centrally located nuclei. Classified as LGMD1A[10,151]
c.179C"/>GS60C50-77SkelSkel: severe MW and wasting. Card: some asymptomic cases; DCM; VEFR; sometimes fatal. Pathology: abnormal and atrophic myofibres with deposits and vacuoles; Myotilin-, CRYAB-, Dystrophin-, Desmin- and Ubiquitin- positive aggregates. Classified as MFM[143,148,152]
c.179C"/>TS60F40-76SkelSkel: difficulty in walking and climbing stairs; MW. Classified as distal myopathy[145]
c.284G"/>TS95I??Pathology: abnormal and atrophic myofibres with deposits and vacuoles; Myotilin-, CRYAB-, Dystrophin-, Desmin- and Ubiquitin- positive aggregates Classified as MFM[143]
c.1214G"/>AR405K41SkelSkel: impossibility to walk long distances; MW. Pathology: abnormal myofibre size; scattered fibres with internally located nuclei; vacuoles and Myotilin-, ZASP-, Desmin- and Actin-positive aggregates. Classified as LGMD1A[142]
c.1214G"/>AR405K41SkelSkel: impossibility to walk long distances; MW. Pathology: abnormal myofibre size; scattered fibres with internally located nuclei; vacuoles and Myotilin-, ZASP-, Desmin- and Actin-positive aggregates. Classified as LGMD1A[140]

Table 4.

Description of clinical and pathological features of myotilinopathies.

[159,161]. The Ig domain containing C-terminus on the other hand is responsible for antiparallel dimerization of Myotilin, which is essential for its function [153,161]. The Myotilin C-terminus alsointeracts directly with Actin, despite the lack of a conventional Actin binding site [162], and is thought to prevent the depolymerisation of Actin filaments and enhance the binding of -Actinin to Actin. Overexpression of Myotilin in CHO cells results in formation of Actin bundles and the delayed expression of Myotilin relative to other Z-disk proteins is thought to be required to avoid premature bundling of Actin fibres [161,162].

Despite the loss of muscle integrity in myotilinopathies Myotilin knockout mice display no muscle defects. Their Z-disk structure and sarcolemma integrity is maintained with no effect on muscle strength and the heart appears normal [163,164]. This suggests that other closely related proteins, such as Palladin and Myopalladin, may have overlapping functions to that of Myotilin and are able to compensate for its loss. Interestingly, mice deficient in Myotilin have a two fold increase in the expression of the muscle stretch sensor Telethonin, which may be responsible for sensing the lack of Myotilin and triggering appropriate signals to prevent muscle failure [163].However, this hypothesis needs to be validated.

Analysis of Myotilin levels in patients compared to control individuals failed to identify a reduction in protein level [10,142] with other studies reporting an increase in Myotilin in some patients [142,165]. This observation leads to the hypothesis that mutations in Myotilin affect its dimerisation or interaction with binding partners, resulting in pathology. This is certainly true in the case of the identified missense mutation affecting the second Ig domain of Myotilin (R405K) resulting in LGMD [142]. The R405K mutation prevents dimerisation disrupting α-Actinin binding and Actin tethering to the Z-disk. As a result, filament stability is lost, the Z-disk is destabilised, and protein aggregates are formed [142]. However, this is not true for the majority of Myotilin mutations, which are found in the serine rich N-terminal domain. In vitro experiments have shown that the S55F, T57I, S60C, and S95I, mutations have no effect on Actin bundling [162], and T57I also has no effect on the interaction with α-Actinin [10]. It has since been hypothesised that mutated Myotilin promotes intermolecular aggregation with other similar Ig domain containing sarcomeric proteins such as Palladin and Titin. The mutant Myotilin expressing transgenic mouse (T57I) that contains aggregates rich in Ig domain containing proteins [151] supports this hypothesis. Interestingly, although in humans the T57I mutation results in LGMD1A in which aggregates are absent, expression in mice results in formation of aggregates that are typical of MFM and SBM. This led to the authors suggesting that the variation in symptoms between MFM, SBM, and LGMD1A, such as protein aggregates, are due to modifying loci [151].

The formation of protein aggregates is a defining feature of MFM and SBM. Aggregate containing muscle from myotilinopathy patients contains increased levels of oxidative stress markers including glycation end products, nitric oxide synthase, superoxide dismutase, and mutant Ubiquitin [166,167]. Protein oxidation promotes protein aggregation and reduces proteolytic degradation. Another factor that may promote protein aggregation in myotilinopathies and other MFMs is the inability of cells to degrade misfolded proteins via the non-lysosomal ubiquitin proteasome system (UPS) responsible for the degradation of 80-90% of myofibrillar proteins [168]. UPS mediated degradation of mutant Myotilin is significantly slower than wildtype Myotilin and inhibition of Myotilin turnover results in aggregates similar to those seen in MFM[169].

Figure 3.

Schematic representation of Myotilin domains and myopathy mutations.

Advertisement

5. ZASP and zaspopathies

Z band alternatively spliced PDZ-containing protein (ZASP)[170], also known as LIM Domain Binding 3 (LDB3), Cypher [171], or Oracle [172] belongs to the PDZ-LIM family of proteins and similar to other members of the family, Enigma [173] and α-Actinin associated LIM protein (ALP) [174], localises to the Z-disk. More than 15 mutations in ZASP have been reported resulting in a range of myopathies including DCM [11,175], HCM [176], MFM [177], inclusion body myositis [178], and LVNCC [11,175,177] (Table 5). ZASP contains a PDZ domain, located at the N-terminus, and an internal ZASP/cypher-like motif (ZM) both capable of interacting with α-Actinin-2 [179-181]. Additionally, the PDZ domain interacts with Myotilin [158] and FATZ[182], which provides structural stability to the Z-disk. The C-terminus contains three LIM domains, which act to recruit signalling proteins to the Z-disk.

As its name suggests the ZASP mRNA is extensively spliced to result in multiple ZASP isoforms, a feature conserved in all species examined with four isoforms in worms [183], 12 isoforms in flies [184-186], 13 in zebrafish [187], and six in mice and humans [11,188] (Figure 4). In mice and humans the isoforms have been characterised according to their length and their expression in the heart or skeletal muscle. So far two short isoforms, (2c, 2s) that lack the LIM domains, and four long isoforms (1c, 1s, 3c and 3s), that contain all three LIM domains, have been characterised [11,170]. Isoforms containing exon four (1s, 2s and 3s) are restricted to cardiac muscle whereas isoforms lacking exon four are found in both cardiac and skeletal muscle [11]. Loss and gain of function experiments have highlighted specific roles for the short and long isoforms. Selective deletion of the short isoforms does not lead to any muscle defects however, loss of the long isoforms results in neonatal lethality in 28% of mice [189]. Surviving knockout mice display growth retardation and Z-disk abnormalities in cardiomyocytes leading to DCM in adulthood, demonstrating the requirement for the

MutationAge of onset /Initial symptomsClinical and pathological features; other studiesRef
c.163G"/>A*V55I?CardCard: LVNCC. Classified as LVNCC[175,191]
c.349G"/>AD117N30-41CardCard: DCM; AVB; EFR; premature death in some cases. Other studies: C2C12 show ZASP aggregates formation and abnormal Actin staining. Classified as LVNCC[11]
c.407C"/>TK136M16CardCard: DCM; VEFR. Classified as LVNCC[11]
c.464G"/>A (mRNA)A147T44-59SkelSkel: progressive MW and MA. Card: some cases of VEFR. Pathology: atrophic, necrotic, and regenerating myofibres; fibre splitting and internally located nuclei; small vacuoles and Desmin-, Myotilin-, CRYAB-, and Dystrophin-positive aggregates; streaming and disintegration of the Z-disk; organelles in clusters. Classified as MFM[177]
c.519C"/>T (mRNA)A165V39-59SkelSkel: progressive MW and MA. Pathology: atrophic, necrotic, and regenerating myofibres; fibre splitting and internally located nuclei; small vacuoles and Desmin-, Myotilin-, CRYAB- and Dystrophin-positive aggregates; streaming and disintegration of the Z-disk; organelles in clusters. Classified as LVNCC[177,192]
c.587C"/>TS196L7-73 (not accurate)CardCard: DCM or HCM; VEFR. Other studies: mice show left ventricular dilation; HCM, VEFR; mild focal fibrosis; sarcomere and Z-disk disorganisation. Classified as DCM[11,176,193]
c.638C"/>TT213I15 monthsCardCard: AVB; VEFR. Other studies: Reduced binding to PGM1 Classified as DCM and LVNCC[11]
c.827C"/>T (mRNA)R268C73SkelSkel: progressive MW. Pathology: atrophic, necrotic, and regenerating myofibres; fibre splitting and internally located nuclei; small vacuoles and Desmin-, Myotilin-, CRYAB-, and Dystrophin-positive aggregates; streaming and disintegration of the Z-disk; organelles in clusters. Classified as MFM[177]
c.1056C"/>G I352M<15-35CardCard: DCM; VEFR. Other studies: Reduced binding to PGM1. Classified as DCM[11]
?D366N68CardCard: HCM. Classified as HCM[176]
?Y468S (+CRSP3 mutation)46CardCard: HCM. Classified as MFM[176,191]
?Q519P21CardCard: HCM. Classified as HCM[176,191]
c.1719G"/>AV566M40SkelSkel: slowly progressive MW; MA. Pathology: abnormal myofibre size; vacuoles and Desmin-, Myotilin-, CRYAB- and Ubiquitin-positive aggregates[178]
?P615L28CardCard: HCM. Classified as HCM[176]
c.1876G"/>AD626Nafter birth-69CardCard: DCM; LVNC. Other studies: mice show that mutant ZASP has higher affinity to PKC, which may cause the heart failure. Classified as LVNCC and DCM[175,191]

Table 5.

Description of clinical and pathological features of zaspopathies.

long, but not the short, isoforms in maintaining Z-disk integrity [189]. Loss of both short and long isoforms however, results in death within the first five days [190] suggesting that there is some redundancy in their roles. This hypothesis is supported by rescue experiments showing that expression of either the short or long skeletal isoform in ZASP deficient mice is sufficient for survival in 19% and 49% of carriers respectively [188]. The different phenotypes observed following loss of long or short isoforms raises the question whether mutations in specific isoforms result in specific myopathies. This is clearly true in the case of mutations in the cardiac specific exon four however, why mutations in exons expressed in both skeletal and cardiac muscle results in only one tissue getting affected is not clear. For example the D117N [11], A147W, and A165V [177] mutations in exon six affect both skeletal and cardiac muscle isoforms. However, D117N preferentially affects the cardiomyocytes whereas A165V preferentially affects the skeletal muscle, and A147W results in both tissues being affected. Therefore, there appears to be no clear correlation between the exon affected and the phenotype presented by the patient.

Examination of the diaphragm muscles, which are not active before birth, in ZASP knockout mice identified little or no difference in the sarcomere structure at E17.5 when compared with wildtype mice, but severe disruption of the Z-disk the day after birth [190], suggesting that ZASP is not required for sarcomere assembly but is critical for maintenance of Z-disk integrity. Examination of cardiac muscle in these mice, which becomes active at E8, at E17.5 identified severely disrupted Z-disks which were completely lost by one day after birth [190]. A role for ZASP in Z-disk maintenance is supported by experiments demonstrating that deletion of ZASP in postnatal hearts results in gradual disruption of the Z-disk and severe DCM resulting in premature death within five months [194]. Targeted deletion of ZASP homologues in Drosophila results in defects in muscle development suggesting a role for ZASP is in Drosophila sarcomerogenesis [184,185]. However, Drosophila has only a single protein equivalent to the mammalian ZASP, ALP, and Enigma proteins. It is therefore possible that in mammals, ZASP, ALP, and Enigma have redundant roles and loss of all three proteins in mammals would result in a phenotype similar to that seen in Drosophila. In Drosophila ZASP was identified as a regulator of cell matrix adhesion localising to integrin adhesion sites in S2 and S2R+ cell lines co-localising with α-Actinin at the Z-disks and integrins at the myotendinous junctions in embryos [184]. ZASP deficient flies display a muscle detachment phenotype and lack α-Actinin at the Z-disk, suggesting that the interaction of ZASP with Integrin is critical in connecting the muscle fibre to the ECM and in directing α–Actinin to the Z-disk [184]. However, localisation of ZASP to myotendinous junctions or costameres has not been reported in any other animal model.

In cardiomyocytes ZASP interacts with Protein Kinase C (PKC) [175], a known modulator of cardiomyocyte growth and contractility. PKC-ε has been shown to interact with RACK-2 and protect cardiomyocytes from ischemic stress [195,196]. Disruption of the PKC-ε - RACK-2 complex results in inhibition of cell contraction [197] and accelerated cell death [198]. In vivo studies have revealed increased levels of PKC in hypertrophy, DCM, and heart failure [199-201], suggesting a role for PKC in stress response, potentially modulated by ZASP. Biochemical analysis of ZASP revealed that the D626N LIM domain mutation increases the binding affinity of ZASP for PKC. The authors suggest that this may reduce the amount of PKC-ε available to bind downstream proteins such as RACK-2 therefore resulting in DCM due to altered distribution of PKC [175]. ZASP also interacts with the metabolic protein Phosphoglucomutase 1 (PGM1), an enzyme involved in glycolysis and gluconeogenesis, through the proline rich regions encoded by exons four, six and, ten and recruits it to the Z-disk [202]. DCM causing mutations in exon four (S196L and T213I) and exon ten (I352M) have been shown to have reduced binding affinity for PGM1 [202]. The binding of ZASP to PGM1 and ZASP mediated targeting of PGM1 to the Z-disk are both increased under stress condition further supporting a role of ZASP in protection and repair of the Z-disk, although the role of PGM1 at the Z-disk is not clear [202].

Figure 4.

Schematic representation of ZASP domains, human splice variants, and mutations.

Advertisement

6. Filamin and filaminopathies

Filamin C (FLNC) was first implicated in MFM in 2005 with the identification of a missense mutation in a German family that presented with weakness of the proximal muscle groups and respiratory insufficiency [12]. Since this initial discovery five additional FLNC mutations have been identified of which two result in MFM [203,204] and three cause distal myopathies in which protein aggregates are not evident [205,206] (Table 6).

MutationAge of onset /Initial symptomsClinical and pathological features; other studiesRef
c.577G"/>AA193T30'sSkelSkel: MW. Pathology: residual fibre size variation; focal increase in fibrosis and internal nuclei. Other studies: C2C12 show increased number of stress fibres and cell projections; FLNC- and Actin-positive aggregates were detected. Classified as distal myopathy[205]
c.752T"/>CM251T30'sSkelSkel: slowly progressive MW. Card: some developed CM. Pathology: abnormal myofibre size; internally located nuclei. Classified as distal myopathy[205]
c.2695_2712del + GTTTGTinsdel(K899_V904) + ins(V899_C900)35-40SkelSkel: progressive MW. Card: AVB. Pathology: variation in myofibre size and increased numbers of internal nuclei; vacuoles and deposits positive for Desmin, Dysferlin, Dystrophin and Ubiquitin; necrotic and regenerating myofibres; nemaline bodies. Classified as MFM[204]
c.2788_2799deldel(V930_T933)34-60SkelSkel: difficulty to stand or walk; progressive MW. Pathology: abnormal myofibre size; atrophic myofibres and internally located nuclei; aggregates positive for FLNC, Ubiquitin, Desmin, Myotilin and CRYAB; nemaline bodies and mitochondria aggregates. Classified as MFM[203]
c.5160delCF1720fsX63320-57SkelSkel: MW and MA. Card: few cases of CM and VEFR. Pathology: from slight myofibre size variation and rare fibre splitting and internally located nuclei to myofibrillar disorganisation, Z-disk streaming, presence of small rods and other deposits. Classified as distal myopathy[206]
c.8130G"/>AW2710X24-49SkelSkel: slowly progressive MW; wheelchair dependence in some patients. Card: some patients with HCM, AVB and VEFR. Pathology: splitting and necrotic fibres; internally located nuclei; aggregates positive for FLNC, Desmin, Myotilin and Dystrophin and vacuoles; Z-disk streaming and nemaline-rod formation. Other studies: protein studies showed a decreased stability and dimerisation capacity of the mutant FLNC; PtK2 cells form aggregates. Classified as MFM[4,12,207]

Table 6.

Description of clinical and pathological features of filaminopathies.

FLNC belongs to the Filamin family of proteins characterised by their ability to cross link Actin. Three Filamin isoforms have been identified all of which are encoded by different genes [208,209]: Filamin A (α-Filamin or Filamin 1) and Filamin B (β-Filamin), which are ubiquitously expressed, and FLNC (Filamin 2, γ-Filamin, Actin Binding Protein 280 (ABP-280) or Actin Binding Protein Ligand (ABP-L)) [210-212], which is expressed specifically in striated and cardiac muscle [204,212]. In striated muscle, FLNC localises in two different pools: 97% of FLNC is contained within the Z-disk of the sarcomere and 3% is found in the sarcolemmal membrane at the level of the costameres and myotendinous junctions [213]. In cardiac muscle FLNC is found in intercalated discs [213,214].

Filamin proteins contain two distinct functional regions (Figure 5). The N-terminal region, which contains two calpain homology domains that are responsible for interacting with Actin and promoting its polymerisation [215]. Two of the three distal myopathy causing FLNC mutations, in which protein aggregates are not evident, are found in this N-terminal region. This suggests that the presence of a functional N-terminal Actin binding domain in MFM causing FLNC mutant protein may be important in the formation of protein aggregates. The other four FLNC mutations are found in the semi-flexible rod domain, which contains 24 homologous Ig-like domains, each about 93 to 103 amino acids long [203]. The Ig-like domains act as an interface for the interaction of FLNC with its binding partners and allow FLNC dimerisation, through domain 24 [216], which is essential for its function.

Disruption of FLNC dimerisation leads to failure of the Z-disk as in the case of W2710X MFM causing FLNC mutation. Truncation of the dimerisation domain results in the loss of secondary structure of the mutant protein hence making it less stable and more susceptible to degradation by proteolytic enzymes [12,207], but is also more prone to aggregation [207]. Although the mutant protein is unable to form dimers, it neither disturbs dimerisation of wildtype FLNC nor affects its interaction with Actin or the sarcoglycans, two key FLNC binding partners [207]. In between FLNC Ig-like domains 15 and 16 (Figure 5), a differentially spliced Hinge 1 (H1) region is present, that provides flexibility to FLNC, but is absent from the predominant form expressed in striated muscle. Additionally, FLNC has a second hinge region (H2, Figure 5) between Ig-like domains 23 and 24, found in both splice variants [210,212], and contains a unique 82 amino acid insert between Ig-like domains 19 and 20 [213], which is thought to recruit FLNC specifically into the Z-disk [157].

Figure 5.

Schematic representation of FLNC domains and mutations.

FLNC has been proposed to have several functions in the muscle. The interaction of Ig-like domain 20 of FLNC with the Z-disk protein Xin is important in regulating the development and remodelling of the Actin cytoskeleton [217]. Additionally, the interaction of FLNC Ig-like domains 19, 20,21, and domain 23 with the Z-disk proteins Myotilin [159] and FATZ [157,182,218,219] maintains the stability of the sarcomere. At the sarcolemma, FLNC interacts with the transmembrane proteins γ- and δ-Sarcoglycans (repeats 20 to 24) [213], Cbl-associated protein (CAP or Ponsin, domain 2) [220], Ankyrin G (repeat 5 and 6) [221], and β1-integrin (domain 20-21) [157]. FLNC therefore connects the Z-disk to the sarcolemma and the ECM providing both a structural linkage and a mechanism for signalling from the sarcolemma to the Z-disk [159,213]. In cardiac muscle FLNC interacts with Nebulette [222], the cardiac specific homologue of the thin filament ruler Nebulin. This interaction has been thought to be important in targeting FLNC to the cytoskeleton therefore ensuring the correct localisation and function of FLNC. FLNC, through Ig-like domains 20, 21, and 23 also interacts with the muscular dystrophy KY protein but the functional importance of this interaction is not known [214]. The identification of a distal myopathy as a result of FLNC haploinsufficiency suggests that the levels of FLNC maybe critical for its function [206]. Additionally, the altered distribution of both sarcomeric and ECM proteins in filaminopathies suggests that the functions of FLNC at the Z-disk and sarcolemma are compromised in filaminopathies. Analysis of the FLNC mouse knockout identified a decrease in the number of primary muscle fibres suggesting a role for FLNC in myogenesis [223]. However, the recent characterisation of a Medaka FLNC mutant showed no difference in the expression of myogenic factors [224]. The role of FLNC in fibre differentiation is therefore still questionable.

The process by which mutations in FLNC result in muscle disease is not understood but the identification of a haploinsufficient form of filaminopathy, and the finding that the W2710X mutant does not disrupt wildtype FLNC dimerisation,together with the severe muscle defects seen in the FLNC knockout mouse suggest that Filamin related MFM manifests as result of direct or indirect loss of functional FLNC. It is therefore hypothesised that the progressive, late-onset, nature of filaminopathies results from a reduction in FLNC function commensurate with the increasing sequestration of wildtype FLNC and FLNC binding partners by mutant FLNC in the cytoplasm.

Advertisement

6. BAG3 and bag3opathies

Bcl2-related athanogene 3 (BAG3, Bis, CAIR) is the most recently identified MFM causing Z-disk protein with the report of a missense mutation (P209L) in exon three resulting in MFM with cardiac complications [13]. Since then 10 additional Bag3 mutations have been reported of which nine result in DCM [225,226] and one in MFM [227] (Table 7). BAG3 is one of six members of the BAG family of proteins. It is a multidomain co-chaperone expressed at high levels in skeletal and cardiac muscle and found at lower levels in tissues such as neurons, adrenal gland, ovaries and testis [228,229]. In skeletal muscle BAG3 co-localises with Desmin and α-Actinin at the Z-disk [228]. An increase in BAG3 expression is detected following an increase in static strain [230], eccentric contraction [231], or nemaline myopathy [232] which suggests that BAG3 plays a rolein repair and regeneration of skeletal muscle injuries caused by mechanical stress and disease.

MutationAge of onset /Initial symptomsClinical and pathological features; other studiesRef
c.211C"/>TR71W41-59CardCard: DCM; VEFR; heart transplantation required. Classified as DCM[225]
c.268C"/>TR90X44CardCard: DCM; VEFR. Classified as DCM[225]
c.326A"/>GH109R21CardCard: DCM; VEFR. Classified as DCM[225]
c.367C"/>TR123X25-36CardCard: DCM in some cases; some cases with VEFR; some required heart transplantation. Classified as DCM[225]
c.626C"/>TP209L5-13Skel and CardSkel: moderate to severe MW and MA; easy fatigability Card: Restrictive HCM; heart transplantation needed in some cases; early death in most cases. Pathology: abnormal myofibre size which larger fibres showed splitting or breakdown; necrotic and regenerating myofibres and internally located nuclei; abnormal fibres with ectopic staining for BAG3, CRYAB, Desmin, Myotilin, Dystrophin and Ubiquitin; presence of dense structures and aggregates of mitochondria; Z-disk streaming. Other studies: neonatal rat cardiomyocytes showed problems in cell fusion; COS-7 cells presented granules in their cytoplasm; C2C12 with reduced BAG3 protein levels show increased apoptosis. Classified as MFM[13,226,228,240]
c.625C"/>T + c.772C"/>TP209W + R258W6SkelSkel: progressed from clumsy walking into MW and decreased spine movement Card: restrictive HCM. Pathology: atrophic fibres; focal myofibrillar disorganisation and degeneration; sarcoplasmic accumulation of granulofilamentous material. Classified as MFM[227]
c.652C"/>TR218W73CardCardiac muscle: ventricular wall thickness; severe VEFR; ectopic atrial rhythm. Other studies: neonatal rat cariomyocyes presented abnormal Z-disk assembly (seen by Desmin and α-Actinin staining) and increased susceptibility to apoptosis. Classified as DCM[226]
c.652CdelR218fsX8947CardCard: DCM; VEFR; early death. Classified as DCM[225]
c.784G"/>AA262T42-44CardCard: DCM; AVB; severe VEFR; required heart transplantation. Classified as DCM[225]
c.1385T"/>CL462P27-34CardCard: ventricular wall thickness; VEFR; cardiac contraction defects. Other studies: neonatal rat cardiomyocytes presented abnormal Z-disk assembly and increased susceptibility to apoptosis. Classified as DCM[226]
c.1430G"/>AA477H47-50CardCard: DCM; severe VEFR; pacemaker insertion. Classified as DCM[225]

Table 7.

Description of clinical and pathological features of bag3opathies.

BAG3 has three recognisable functional domains (Figure 6); no mutations have been reported in the WW domain containing N-terminal region, which interacts with proline rich motifs of signal transduction proteins, or in the proline rich central region, which interacts with WW domains and Src3 homology (SH3) domains of signal transduction proteins such as Phospholipase C (PLCγ) [233].

Two mutations have however been reported in the evolutionary conserved C-terminal BAG domain that has a key role in the apoptotic and chaperone functions of BAG3 [234-236]. The BAG domain binds with high affinity to, and regulates, stress inducible Heat shock protein 70 (Hsp70) [233-235,237] and the constitutively expressed Heatshock cognate protein 70 (Hsc70) [237], that ensure

correct protein folding and targeting of misfolded proteins for proteasomal degradation [238]. BAG3 competitively binds to the ATPase domain of these chaperone proteins and alters their chaperone properties thereby targeting chaperone associated proteins for proteasomal degradation [237,239]. In fact, BAG3 has recently been shown to form a stable complex with the small Heat shock protein HspB8 and stimulate macroautophagy [236], a process that is particularly important in Huntington disease where association of BAG3 with HspB8 promotes degradation of mutant Huntingtin [236,241]. In inclusion body myositis, macroautophagy plays a role in removing β-amyloid aggregates [242] and it is possible that BAG3 also plays a role in this cellular response to protein aggregates in MFM. However, as the primary defect in MFM is the dissolution of muscle fibres beginning at the Z-disk preventing the formation of protein aggregates is unlikely to be sufficient to prevent muscle pathology.

In B-crystallinopathies BAG3 suppresses protein aggregation and toxicity by preferentially binding mutant CRYAB, reducing its aggregation and increasing its solubility [136]. This demonstrates that BAG3 not only indirectly regulates protein folding and degradation but also has the potential to prevent misfolding and promote degradation of mutant proteins thereby preventing disease pathology. Interestingly, deletion of the BAG domain results in a similar inhibition of aggregation of mutant CRYAB. BAG3 may therefore function through a pathway that is independent of Hsp70/Hsc70 [136]. BAG3 synergisticallyinteracts with Bcl-2, via the BAG domain, to prevent Bax induced and FasL-

Fas mediated apoptosis [239,243]. BAG3 levels are up-regulated in response to oxidative stress [244], heat shock, heavy metal exposure [245,246], or photoinjury in the retina [247] and increased levels of BAG3 in human epithelial cells has been show to result in decreased Bax or Fas mediated apoptosis demonstrating the critical adaptive role of BAG3 in response to cell stress.

Immunohistochemistry on P209L mutant muscle biopsy samples revealed increased immunoreactivity in abnormal fibres for the chaperone molecules Hsp27 and CRYAB and the anti- apoptotic protein Bcl-2. This was accompanied by increased apoptosis suggesting that the P209L mutation interfered with the anti-apoptotic functions of BAG3 [13]. Non-denaturing gel electrophoresis revealed faster migration of the mutant BAG3 complex than wildtype [13], suggesting that the loss of function may be due to reduced interaction with partner proteins, possibly Bcl-2, given that an increase in apoptosis is observed. This is supported by transfection of P209L mutant BAG3 into neonatal cardiomyocytes resulting in increased susceptibility to stress mediated apoptosis [226] and the observation that mice deficient in BAG3 also display increased apoptosis [228]. It has been shown that the down-regulation of BAG3 enhances the apoptotic response to chemotherapy in lymphocytic leukaemia cells making it a potential target for cancer therapies [248], and further demonstrating its anti-apoptotic role.

Since the primary defect in BAG3opathies is the fragmentation of fibres it can be postulated that perhaps BAG3 has a role in muscle development or maintenance of muscle structure. BAG3 deficient mice are normal at birth but cease to gain weight at day 12. Muscle histology revealed myofibril and Z-disk defects with no sarcolemma damage [228]. Taken together, this data suggests that BAG3 is not necessary for sarcomerogenesis but is critical for maintenance of fibre integrity. By the 25th day BAG3 deficient mice die as a result of intercostal muscle failure or pulmonary oedema that results in cardiac failure [228]. Targeted knockdown of BAG3 in zebrafish has also resulted in severe cardiac defects demonstrating a critical role of BAG3 in maintaining the structural integrity of cardiomyocytes [225]. A recent study has shown that BAG3 regulates myofibril stability by facilitating the interaction of Hsc70 with CapZ, a protein that caps the barbed ends of Actin filaments that extend into Z-disk [230]. Loss of BAG3 makes CapZ more vulnerable to degradation resulting in loss of CapZ and fibre fragmentation following mechanical stress [230].

Figure 6.

Schematic representation BAG3 domains and mutations.

In summary, the most recently identified MFM causing gene plays key roles in the localisation of CapZ to the Z-disk through its interaction with Hsc70, protein folding and degradation, and in the regulation of apoptosis. Given the indirect role BAG3 plays in Z-disk and muscle function, its binding partners are excellent candidates for further genes that may be mutated in MFM. However, given the fact that most of the identified disease causing mutations lie outside a recognised domain in BAG3 there may be many more functions for BAG3 that remain to be characterised, perhaps including a direct role in the Z-disk.

Advertisement

7. Conclusion

The many functions of the MFM proteins, which are themselves just a very small subset of the Z-disk associated proteins, highlights the complex and dynamic nature of the Z-disk. Whilst a characteristic feature of MFM is dissolution of the myofibril, originating at the Z-disk, these disorders are not due to simple loss or disruption of structural components in this tensile load bearing structure. This is exemplified by the identification of BAG3, which localises to the Z-disk but appears to only have indirect association to it, as an MFM protein. Further support comes from analysis of mouse knockouts for the MFM genes, none of which have defects in the formation of myofibrils.

The progressive nature of the disease and the identification of roles for the associated proteins in muscle repair and maintenance is more suggestive of a gradual accumulation of defects in Z-disk organisation eventually leading to structural failure. An interesting finding from mice lacking the long isoforms of ZASP is that they have increased levels of the MFM proteins Myotilin, CRYAB, and FLNC as well as the extracellular matrix proteins β1D Integrin and the sarcoglycans [189]. Up-regulation of Z-disk components is observed in α-Actinin-3 knockout mice in which FLNC, Myotilin, ZASP, and CRYAB are up-regulated [249]. Increased levels of FLNC are also detected in patients with LGMD or Duchenne muscular dystrophy [213]. This data, together with that previously presented, strongly supports the idea that the MFM proteins are up-regulated to protect the sarcomere and ECM from damage, whether that damage is caused by muscle activity, mutation of muscle proteins, or increases in oxidative, metabolic, and other forms of cell stress. Whether this is through a general stress response pathway that up-regulates the expression of Z-disk associated proteins or through a more specific pathway that selectively target proteins based on the nature of the stress remains to be determined.

For more than half of the cases of MFM the causative mutation is not known. As we have described there are many binding partners for the known MFM proteins, mutations in which may account for some of these cases. Additionally there is evidence from experiments with Desmin that mutations in other genes may act as modifiers of disease. Far more mutations have been identified in Desmin than in other MFM genes and it may be that modifiers will be identified in other subtypes of MFM as larger cohorts are analysed. Given the hypothesised role for the MFM proteins in stress response it is possible that any mutations that result in cellular stress may modify the presentation of MFM perhaps accounting for some of the differences in age of onset. Furthermore, differences in stress between cardiac and skeletal tissues may explain the differences in symptoms between these tissues, even in individuals with the same mutation. As the application of whole genome sequencing to mutation detection in myopathy becomes more widespread it may be possible to identify potential modifiers and investigate their role in MFM.

The existing literature on MFM and the MFM associated proteins has identified many exciting avenues for investigation. To investigate these areas further animal models, modelling specific MFM mutations, are required that would allow for better characterisation of pathology and the progression of disease together with a consistent genetic background to allow the analysis of potential genetic modifiers. The development of better tools to investigate the function of the MFM proteins, together with the identification of further MFM genes and modifiers, will allow us to improve our understanding of the many diverse and complex roles of these Z-disk associated proteins and move closer to the development of effective therapies for these conditions.

8. Abbreviations

ACD domain: -crystallin domain
AVB: atrioventricular block
BAG3: Bcl2-related athanogene 3
CM: cardiomyopathy
CRYAA: αA-Crystallin
CRYAB: αB-Crystallin
DCM: dilated cardiomyopathy
DRM: Desmin-related myopathy
ECM: extracellular matrix
FLNC: Filamin C
HCM: hypertrophic cardiomyopathy
IF: intermediate filament
Ig: immunoglobulin
LVNCC: left ventricular non-compaction cardiomyopathy
MA: muscle atrophy
MFM: myofibrillar myopathy
MW: muscle weakness
PGM1: Phosphoglucomutase 1
PKC: Protein kinase C
SBM: spheroid body myopathy
ULF: unit length filament
UPS: ubiquitin proteasome system
UTR: untranslated region
VEFR: ventricular ejection fraction reduction
ZASP: Z-band alternatively spliced PDZ-motif protein
WT: wildtype
ZM: ZASP/Cypher-like motif
GFAP: Glial fibrillary acidic protein

Acknowledgement

The authors would like to thank Dr Jihane Homman-Ludiye for her comments on the manuscript.

References

  1. 1. NakanoS.EngelA. G.WaclawikA. J.Emslie-SmithA. M.BusisN. A.Myofibrillar myopathy with abnormal foci of desmin positivity. I. Light and electron microscopy analysis of 10 cases. J. Neuropathol. Exp. Neurol. 19965554962
  2. 2. OlivéM.OdgerelZ.MartínezA.PozaJ. J.BragadoF. G.ZabalzaR. J.JericóI.Gonzalez-MeraL.ShatunovA.Lee-SH.ArmstrongJ.MaravíE.ArroyoM. R.Pascual-CalvetJ.NavarroC.ParadasC.HuertaM.MarquezF.RivasE. G.PouA.FerrerI.GoldfarbL. G.Clinical and myopathological evaluation of early- and late-onset subtypes of myofibrillar myopathy. Neuromuscul Disord 2011
  3. 3. ClaeysK. G.VenP. F. M.BehinA.StojkovicT.EymardB.DubourgO.LaforêtP.FaulknerG.RichardP.VicartP.RomeroN. B.StoltenburgG.UddB.FardeauM.VoitT.DOFürstDifferential involvement of sarcomeric proteins in myofibrillar myopathies: a morphological and immunohistochemical study. Acta Neuropathol. 2009117293307
  4. 4. KleyR. A.HellenbroichY.van der VenP. F. M.DOFurstHuebner. A.BruchertseiferV.PetersS. A.HeyerC. M.KirschnerJ.SchroderR.FischerD.MullerK.TolksdorfK.EgerK.GermingA.BrodherrT.ReumC.WalterM. C.LochmullerH.KetelsenU. P.VorgerdM.Clinical and morphological phenotype of the filamin myopathy: a study of 31 German patients. Brain 2007130325064
  5. 5. SchröderR.SchoserB.MyofibrillarMyopathies. A.ClinicalMyopathologicalGuide.Brain Pathology 20091948392
  6. 6. FerrerI.OlivéM.Molecular pathology of myofibrillar myopathies. Expert Reviews in Molecular Medicine 2008
  7. 7. SelcenD.Myofibrillarmyopathies.NeuromusculDisord 20112116171
  8. 8. HorowitzS. H.SchmalbruchH.Autosomal dominant distal myopathy with desmin storage: a clinicopathologic and electrophysiologic study of a large kinship. Muscle Nerve 19941715160
  9. 9. LacsonA. G.SeshiaS. S.SarnatH. B.AndersonJ.De GrootW. R.ChudleyA.AdamsC.DarwishH. Z.LowryR. B.KuhnS.Autosomalrecessive.fatalinfantile.hypertonicmuscular.dystrophyamong.CanadianNatives.Canadian Natives. Can J Neurol Sci 19942120312
  10. 10. HauserM.HorriganS.SalmikangasP.TorianU.VilesK.DancelR.TimR.TaivainenA.BartoloniL.GilchristJ.StajichJ.GaskellP.GilbertJ.VanceJ.Pericak-VanceM.CarpénO.WestbrookC.SpeerM.Myotilin is mutated in limb girdle muscular dystrophy 1A. Hum. Mol. Genet. 2000921417
  11. 11. VattaM.MohapatraB.JimenezS.SanchezX.FaulknerG.PerlesZ.SinagraG.LinJ. H.VuT. M.ZhouQ.BowlesK. R.Di LenardaA.SchimmentiL.FoxM.MAChriscoMurphy. R. T.Mc KennaW.ElliottP.BowlesN. E.ChenJ.ValleG.TowbinJ. A.Mutationsin.CypherZ. A. S. P.inpatients.withdilated.cardiomyopathyleftventricular.non-compactionJ. Am. Coll. Cardiol. 200342201427
  12. 12. VorgerdM.van der VenP. F. M.BruchertseiferV.LöweT.KleyR. A.SchröderR.LochmüllerH.HimmelM.KoehlerK.DOFürstHuebner. A.A, Schröder R, Lochmüller H, Himmel M, Koehler K, Fürst DO, Huebner A. A mutation in the dimerization domain of filamin c causes a novel type of autosomal dominant myofibrillar myopathy. Am. J. Hum. Genet. 200577297304
  13. 13. SelcenD.MuntoniF.BurtonB. K.PegoraroE.SewryC.BiteA. V.EngelA. G.Mutationin. B. A. G.causessevere.dominantchildhood.musculardystrophy.Ann. Neurol. 200965839
  14. 14. SelcenD.EngelA. G.MyofibrillarMyopathy.SeattleGene.GeneReviews; 2005
  15. 15. ClaeysK. G.FardeauM.SchroderR.SuominenT.TolksdorfK.BehinA.DubourgO.EymardB.MaisonobeT.StojkovicT.FaulknerG.RichardP.VicartP.UddB.VoitT.StoltenburgG.Electron microscopy in myofibrillar myopathies reveals clues to the mutated gene. Neuromuscul Disord 20081865666
  16. 16. LazaridesE.BDHubbardImmunological characterization of the subunit of the 100 A filaments from muscle cells. Proc. Natl. Acad. Sci. U.S.A. 19767343448
  17. 17. Price MG.Molecular analysis of intermediate filament cytoskeleton--a putative load-bearing structure. Am. J. Physiol. 1984H56672
  18. 18. SchroderR.DOFurstKlasen. C.ReimannJ.HerrmannH.van der VenP. F.Association of plectin with Z-discs is a prerequisite for the formation of the intermyofibrillar desmin cytoskeleton. Lab Invest 20008045564
  19. 19. BennardiniF.WrzosekA.ChiesiM.Alpha B-crystallin in cardiac tissue. Association with actin and desmin filaments. Circ. Res. 19927128894
  20. 20. Bang-LM.GregorioC.LabeitS.Molecular dissection of the interaction of desmin with the C-terminal region of nebulin. J. Struct. Biol. 200213711927
  21. 21. Conover GM, Gregorio CC.The desmin coil 1B mutation K190A impairs nebulin Z-disc assembly and destabilizes actin thin filaments. J Cell Sci 2011124346476
  22. 22. Bellin RM, Huiatt TW, Critchley DR, Robson RM.Synemin may function to directly link muscle cell intermediate filaments to both myofibrillar Z-lines and costameres. J Biol Chem 2001276323307
  23. 23. BrecklerJ.LazaridesE.Isolation of a new high molecular weight protein associated with desmin and vimentin filaments from avian embryonic skeletal muscle. J. Cell Biol. 198292795806
  24. 24. MizunoY.ThompsonT. G.GuyonJ. R.LidovH. G.BrosiusM.ImamuraM.OzawaE.WatkinsS. C.KunkelL. M.Desmuslinan.intermediatefilament.proteinthat.interactswith.alpha-dystrobrevindesminProc. Natl. Acad. Sci. U.S.A. 200198615661
  25. 25. CartaudA.JasminB. J.ChangeuxJ. P.CartaudJ.Direct involvement of a lamin-B-related (54 kDa) protein in the association of intermediate filaments with the postsynaptic membrane of the Torpedo marmorata electrocyte. J Cell Sci 1995Pt 1):153-60.
  26. 26. FavreB.SchneiderY.LingasamyP.Bouameur-EJ.BegréN.GontierY.Steiner-Champliaud-FM.MAFriasBorradori. L.FontaoL.Plectin interacts with the rod domain of type III intermediate filament proteins desmin and vimentin. Eur. J. Cell Biol. 201190390400
  27. 27. SteinertP. M.ChouY. H.PrahladV.ParryD. A.MarekovL. N.WuK. C.JangS. I.GoldmanR. D. A.highmolecular.weightintermediate.filament-associatedprotein.inB. H.K-cellsis.nestina.typeV. I.intermediatefilament.proteinLimited co-assembly in vitro to form heteropolymers with type III vimentin and type IV alpha-internexin. J Biol Chem 1999274988190
  28. 28. Langley RC, Cohen CM.Association of spectrin with desmin intermediate filaments. J. Cell. Biochem. 1986301019
  29. 29. GeorgatosS. D.WeberK.GeislerN.BlobelG.Binding of two desmin derivatives to the plasma membrane and the nuclear envelope of avian erythrocytes: evidence for a conserved site-specificity in intermediate filament-membrane interactions. Proc. Natl. Acad. Sci. U.S.A. 19878467804
  30. 30. ThornellL.CarlssonL.LiZ.MericskayM.PaulinD.Null mutation in the desmin gene gives rise to a cardiomyopathy. J. Mol. Cell. Cardiol. 199729210724
  31. 31. HerrmannH.AebiU.Intermediate filaments and their associates: multi-talented structural elements specifying cytoarchitecture and cytodynamics. Curr. Opin. Cell Biol. 2000127990
  32. 32. WeberK.GeislerN.Intermediatefilaments.structuralconservation.divergenceAnn. N. Y. Acad. Sci. 198545512643
  33. 33. FuchsE.WeberK.Intermediatefilaments.structuredynamics.functiondiseaseAnnu.RevBiochem. 19946334582
  34. 34. BrownJ. H.CohenC.ParryD. A.Heptad breaks in alpha-helical coiled coils: stutters and stammers. Proteins 19962613445
  35. 35. GoldfarbL. G.OlivéM.VicartP.GoebelH. H.Intermediatefilament.diseasesdesminopathy.AdvExp.MedBiol. 200864213164
  36. 36. SelcenD.Myofibrillarmyopathy.clinicalmorphological.geneticstudies.in.patientsBrain 200412743951
  37. 37. SharmaS.MückeN.KatusH. A.HerrmannH.BärH.Disease mutations in the “head” domain of the extra-sarcomeric protein desmin distinctly alter its assembly and network-forming properties. J. Mol. Med. 200987120719
  38. 38. WahbiK.BehinA.CharronP.DunandM.RichardP.MeuneC.VicartP.LaforêtP.StojkovicT.BécaneH. M.KuntzerT.DubocD.High cardiovascular morbidity and mortality in myofibrillar myopathies due to DES gene mutations: a 10-year longitudinal study. Neuromuscul Disord 2012222118
  39. 39. BärH.GoudeauB.WäldeS.Casteras-SimonM.MückeN.ShatunovA.GoldbergY. P.ClarkeC.HoltonJ. L.EymardB.KatusH. A.FardeauM.GoldfarbL.VicartP.HerrmannH.Conspicuous involvement of desmin tail mutations in diverse cardiac and skeletal myopathies. Hum. Mutat. 20072837486
  40. 40. BärH.SchopfererM.SharmaS.HochsteinB.MückeN.HerrmannH.WillenbacherN.Mutations in desmin’s carboxy-terminal’tail’ domain severely modify filament and network mechanics. J. Mol. Biol. 2010397118898
  41. 41. BergmanJ. E. H.Veenstra-KnolH. E.van EssenA. J.van RavenswaaijC. M. A.Dunnenden. W. F. A.van denWijngaard. A.Peter vanTintelen. J.Two related Dutch families with a clinically variable presentation of cardioskeletal myopathy caused by a novel S13F mutation in the desmin gene. European Journal of Medical Genetics 20075035566
  42. 42. PicaE. C.KathirvelP.PramonoZ. A. D.Lai-SP.Yee-CW.Characterization of a novel S13F desmin mutation associated with desmin myopathy and heart block in a Chinese family. Neuromuscul Disord 20081817882
  43. 43. Spaendonck-ZwartsK. Y.KooiA. J.BergM. P.IppelE. F.BovenL. G.YeeW. C.WijngaardA.BrusseE.HoogendijkJ. E.DoevendansP. A.VisserM.JongbloedJ. D. H.TintelenJ. P.Recurrent and founder mutations in the Netherlands: the cardiac phenotype of DES founder mutations S13Fand p.N342D. Neth Heart J 2011
  44. 44. ArbustiniE.PasottiM.PilottoA.PellegriniC.GrassoM.PrevitaliS.RepettoA.BelliniO.AzanG.ScaffinoM.Desmin accumulation restrictive cardiomyopathy and atrioventricular block associated with desmin gene defects. European Journal of Heart Failure 2006847783
  45. 45. TaylorM. R. G.SlavovD.KuL.Di LenardaA.SinagraG.CarnielE.HauboldK.MMBoucekFerguson. D.GrawS. L.ZhuX.CavanaughJ.SucharovC. C.LongC. S.BristowM. R.LavoriP.MestroniL.forthe.FamilialCardiomyopathy.RegistrytheB. E. S. T. .Beta-BlockerEvaluation.ofSurvival.TrialD. N. A.BankPrevalence of Desmin Mutations in Dilated Cardiomyopathy. Circulation 2007
  46. 46. HeY.ZhangZ.HongD.DaiQ.JiangT.Myocardial fibrosis in desmin-related hypertrophic cardiomyopathy. Journal of Cardiovascular Magnetic Resonance 2010
  47. 47. ArizaA.CollJ.Fernández-FiguerasM. T.MDLópezMate. J. L.GarcíaO.Fernández-VasaloA.Navas-PalaciosJ. J.Desminmyopathy. a.multisystemdisorder.involvingskeletal.cardiacsmoothmuscle.Hum. Pathol. 19952610327
  48. 48. Muñoz-MármolA. M.StrasserG.IsamatM.CoulombeP. A.YangY.RocaX.VelaE.MateJ. L.CollJ.Fernández-FiguerasM. T.Navas-PalaciosJ. J.ArizaA.FuchsE. A.dysfunctionaldesmin.mutationin. a.patientwith.severegeneralized.myopathyProc. Natl. Acad. Sci. U.S.A. 199895113127
  49. 49. BärH.MückeN.KostarevaA.SjöbergG.AebiU.HerrmannH.Severe muscle disease-causing desmin mutations interfere with in vitro filament assembly at distinct stages. Proc. Natl. Acad. Sci. U.S.A. 200510215099104
  50. 50. GoudeauB.Rodrigues-LimaF.FischerD.Casteras-SimonM.SambuughinN.de VisserM.LaforêtP.FerrerX.ChaponF.SjöbergG.KostarevaA.SejersenT.DalakasM. C.GoldfarbL. G.VicartP.Variable pathogenic potentials of mutations located in the desmin alpha-helical domain. Hum. Mutat. 20062790613
  51. 51. ParkK. Y.DalakasM. C.GoebelH. H.FerransV. J.Semino-MoraC.LitvakS.TakedaK.GoldfarbL. G.Desmin splice variants causing cardiac and skeletal myopathy. J. Med. Genet. 2000378517
  52. 52. DalakasM. C.ParkK. Y.Semino-MoraC.LeeH. S.SivakumarK.GoldfarbL. G.Desminmyopathy. a.skeletalmyopathy.withcardiomyopathy.causedby.mutationsin.thedesmin.geneN. Engl. J. Med. 200034277080
  53. 53. ClemenC. S.FischerD.ReimannJ.EichingerL. M.llerC. R. M.llerH. D.GoebelH. H.Schr derR.How much mutant protein is needed to cause a protein aggregate myopathy in vivo? Lessons from an exceptional desminopathy. Hum. Mutat. 2009E4909
  54. 54. SchroderR.GoudeauB.SimonM.FischerD.EggermannT.ClemenC.LiZ.ReimannJ.XueZ.Rudnik-SchonebornS.ZerresK.van der VenP.FurstD.KunzW.VicartP.On noxious desmin: functional effects of a novel heterozygous desmin insertion mutation on the extrasarcomeric desmin cytoskeleton and mitochondria. Hum. Mol. Genet. 20031265769
  55. 55. VrabieA.GoldfarbL. G.ShatunovA.NägeleA.FritzP.KaczmarekI.GoebelH. H.The enlarging spectrum of desminopathies: new morphological findings, eastward geographic spread, novel exon 3 desmin mutation. Acta Neuropathol. 20051094117
  56. 56. GoldfarbL. G.ParkK. Y.CervenákováL.GorokhovaS.LeeH. S.VasconcelosO.NagleJ. W.Semino-MoraC.SivakumarK.DalakasM. C.Missense mutations in desmin associated with familial cardiac and skeletal myopathy. Nat. Genet. 1998194023
  57. 57. YuriT.MikiK.TsukamotoR.ShindeA.KusakaH.TsuburaA.Autopsy case of desminopathy involving skeletal and cardiac muscle. Pathol Int 200757326
  58. 58. SjöbergG.CASaavedra-MatizRosen. D. R.WijsmanE. M.BorgK.HorowitzS. H.SejersenT. A.missensemutation.inthe.desminrod.domainis.associatedwith.autosomaldominant.distalmyopathy.exertsa.dominantnegative.effecton.filamentformation.Hum. Mol. Genet. 1999821918
  59. 59. CarlssonL.FischerC.SjöbergG.RobsonR. M.SejersenT.Thornell-EL.Cytoskeletal derangements in hereditary myopathy with a desmin L345P mutation. Acta Neuropathol. 2002104493504
  60. 60. BärH.FischerD.GoudeauB.KleyR. A.ClemenC. S.VicartP.HerrmannH.VorgerdM.SchröderR.Pathogenic effects of a novel heterozygous R350P desmin mutation on the assembly of desmin intermediate filaments in vivo and in vitro. Hum. Mol. Genet. 200514125160
  61. 61. FidzianskaA.KotowiczJ.SadowskaM.GoudeauB.WalczakE.VicartP.Hausmanowa-PetrusewiczI. A.noveldesmin.R3mutationP.causescardiac.skeletalmyopathy.Neuromuscul Disord 20051552531
  62. 62. DagvadorjA.GoudeauB.Hilton-JonesD.BlancatoJ. K.ShatunovA.Simon-CasterasM.SquierW.NagleJ. W.GoldfarbL. G.VicartP.Respiratory insufficiency in desminopathy patients caused by introduction of proline residues in desmin c-terminal alpha-helical segment. Muscle Nerve 20032766975
  63. 63. KaminskaA.StrelkovS. V.GoudeauB.OlivéM.DagvadorjA.FidzianskaA.Simon-CasterasM.ShatunovA.DalakasM. C.FerrerI.KwiecinskiH.VicartP.GoldfarbL. G.Small deletions disturb desmin architecture leading to breakdown of muscle cells and development of skeletal or cardioskeletal myopathy. Hum. Genet. 200411430613
  64. 64. OlivéM.GoldfarbL.MorenoD.LaforetE.DagvadorjA.SambuughinN.Martínez-MatosJ. A.MartínezF.AlióJ.FarreroE.VicartP.FerrerI.Desmin-relatedmyopathy.clinicalelectrophysiological.radiologicalneuropathological.geneticstudies.J. Neurol. Sci. 200421912537
  65. 65. OlivéM.ArmstrongJ.MirallesF.PouA.FardeauM.GonzalezL.MartínezF.FischerD.Martínez-MatosJ. A.ShatunovA.GoldfarbL.FerrerI.Phenotypic patterns of desminopathy associated with three novel mutations in the desmin gene. Neuromuscul Disord 20071744350
  66. 66. AriasM.PardoJ.Blanco-AriasP.Sobrido-JM.AriasS.DapenaD.CarracedoÁ.GoldfarbL. G.NavarroC.Distinct phenotypic features and gender-specific disease manifestations in a Spanish family with desmin L370P mutation. Neuromuscul Disord 200616498503
  67. 67. StrachK.SommerT.GrohéC.MeyerC.FischerD.WalterM. C.VorgerdM.ReilichP.BärH.ReimannJ.ReunerU.GermingA.GoebelH. H.LochmüllerH.WinterspergerB.SchröderR.Clinicalgenetic.cardiacmagnetic.resonanceimaging.findingsin.primarydesminopathies.Neuromuscul Disord 20081847582
  68. 68. SugawaraM.KatoK.KomatsuM.WadaC.KawamuraK.ShindoS.YoshiokaN.TanakaK.WatanabeS.ToyoshimaI. A.novel denovo.mutationin.thedesmin.genecauses.desminmyopathy.withtoxic.aggregatesNeurology 20005598690
  69. 69. GoudeauB.DagvadorjA.Rodrigues-LimaF.NédellecP.Casteras-SimonM.PerretE.LangloisS.GoldfarbL.VicartP.Structural and functional analysis of a new desmin variant causing desmin-related myopathy. Hum. Mutat. 20011838896
  70. 70. DagvadorjA.OlivM.Urtizberea-AJ.HalleM.ShatunovA. B.nnemannC.Park-YK.GoebelH. H.FerrerI.VicartP.DalakasM. C.GoldfarbL. G. A.seriesof.WestEuropean.patientswith.severecardiac.skeletalmyopathy.associatedwith. a.de novoR4.mutationW.indesmin.J. Neurol. 20042511439
  71. 71. PruszczykP.Kostera-PruszczykA.ShatunovA.GoudeauB.DramiñskaA.TakedaK.SambuughinN.VicartP.StrelkovS. V.GoldfarbL. G.KaminskaA.Restrictive cardiomyopathy with atrioventricular conduction block resulting from a desmin mutation. International Journal of Cardiology 200711724453
  72. 72. LiD.TapscoftT.GonzalezO.BurchP. E.MAQuiñonesZoghbi. W. A.HillR.BachinskiL. L.MannD. L.RobertsR.Desmin mutation responsible for idiopathic dilated cardiomyopathy. Circulation 19991004614
  73. 73. MiyamotoY.Frequency and clinical characteristics of dilated cardiomyopathy caused by desmin gene mutation in a Japanese population. European Heart Journal 20012222849
  74. 74. DalakasM. C.DagvadorjA.GoudeauB.Park-YK.TakedaK.Simon-CasterasM.VasconcelosO.SambuughinN.ShatunovA.NagleJ. W.SivakumarK.VicartP.GoldfarbL. G.Progressiveskeletal.myopathya.phenotypicvariant.ofdesmin.myopathyassociated.withdesmin.mutationsNeuromuscul Disord 2003132528
  75. 75. MuntoniF.Disease severity in dominant Emery Dreifuss is increased by mutations in both emerin and desmin proteins. Brain 200612912608
  76. 76. ConoverG. M.HendersonS. N.GregorioC. C. A.myopathy-linkeddesmin.mutationperturbs.striatedmuscle.actinfilament.architectureMol Biol Cell 20092083445
  77. 77. LiZ.Colucci-GuyonE.Pinçon-RaymondM.MericskayM.PourninS.PaulinD.BabinetC.Cardiovascular lesions and skeletal myopathy in mice lacking desmin. Dev. Biol. 19961753626
  78. 78. MilnerD. J.WeitzerG.TranD.BradleyA.CapetanakiY.Disruption of muscle architecture and myocardial degeneration in mice lacking desmin. J. Cell Biol. 1996134125570
  79. 79. BaloghJ.MerisckayM.LiZ.PaulinD.ArnerA.Hearts from mice lacking desmin have a myopathy with impaired active force generation and unaltered wall compliance. Cardiovasc. Res. 20025343950
  80. 80. CapetanakiY.Desmincytoskeleton. a.potentialregulator.ofmuscle.mitochondrialbehavior.functionTrends Cardiovasc. Med. 20021233948
  81. 81. MilnerD. J.MavroidisM.WeislederN.CapetanakiY.Desmin cytoskeleton linked to muscle mitochondrial distribution and respiratory function. J. Cell Biol. 2000150128398
  82. 82. LiZ.MericskayM.AgbulutO.Butler-BrowneG.CarlssonL.ThornellL. E.BabinetC.PaulinD.Desmin is essential for the tensile strength and integrity of myofibrils but not for myogenic commitment, differentiation, and fusion of skeletal muscle. J. Cell Biol. 199713912944
  83. 83. WangX.OsinskaH.DornG. W.NiemanM.LorenzJ. N.GerdesA. M.WittS.KimballT.GulickJ.RobbinsJ.Mouse model of desmin-related cardiomyopathy. Circulation 200110324027
  84. 84. GroenenP. J.MerckK. B.de JongW. W.BloemendalH.Structure and modifications of the junior chaperone alpha-crystallin. From lens transparency to molecular pathology. Eur. J. Biochem. 1994225119
  85. 85. IwakiT.Kume-IwakiA.LiemR. K.GoldmanJ. E.Alpha B-crystallin is expressed in non-lenticular tissues and accumulates in Alexander’s disease brain. Cell 198957718
  86. 86. BhatS. P.NagineniC. N.alphaB.subunitof.lens-specificprotein.alpha-crystallinis.presentin.otherocular.non-oculartissues.Biochem Biophys Res Commun 198915831925
  87. 87. LongoniS.JamesP.ChiesiM.Cardiacalpha-crystallin. I.IsolationidentificationMol.CellBiochem. 19909911320
  88. 88. AtomiY.ToroK.MasudaT.HattaH.Fiber-type-specific alphaB-crystallin distribution and its shifts with T(3) and PTU treatments in rat hindlimb muscles. J. Appl. Physiol. 200088135564
  89. 89. LeachI. H.TsangM. L.ChurchR. J.LoweJ.Alpha-Bcrystallin.inthe.normalhuman.myocardiumcardiacconducting.systemJ. Alpha-B crystallin in the normal human myocardium and cardiac conducting system. J. Pathol. 199417325560
  90. 90. DjabaliK.de NéchaudB.LandonF.MMPortierAlpha.AlphaB-crystallin interacts with intermediate filaments in response to stress. J Cell Sci 1997Pt 21):2759-69.
  91. 91. Nicholl ID, Quinlan RA.Chaperone activity of alpha-crystallins modulates intermediate filament assembly. Embo J. 19941394553
  92. 92. PlaterM. L.GoodeD.MJCrabbeEffects of site-directed mutations on the chaperone-like activity of alphaB-crystallin. J Biol Chem 19962712855866
  93. 93. HorwitzJ.Alpha-crystallin can function as a molecular chaperone. Proc. Natl. Acad. Sci. U.S.A. 1992891044953
  94. 94. RamanB.RaoC. M.Chaperone-like activity and quaternary structure of alpha-crystallin. J Biol Chem 1994269272648
  95. 95. BerengianA. R.ParfenovaM.MchaourabH. S.Site-directed spin labeling study of subunit interactions in the alpha-crystallin domain of small heat-shock proteins. Comparison of the oligomer symmetry in alphaA-crystallin, HSP 27, and HSP 16.3. J Biol Chem 1999274630514
  96. 96. HeadM. W.CorbinE.GoldmanJ. E.Coordinate and independent regulation of alpha B-crystallin and hsp27 expression in response to physiological stress. J. Cell. Physiol. 19941594150
  97. 97. ClarkA. R.CENaylorBagnéris. C.KeepN. H.SlingsbyC.Crystal structure of R120G disease mutant of human αB-crystallin domain dimer shows closure of a groove. J. Mol. Biol. 201140811834
  98. 98. BovaM. P.YaronO.HuangQ.DingL.HaleyD. A.StewartP. L.HorwitzJ.MutationR1.inG.alphaB-crystallin.whichis.linkedto. a.desmin-relatedmyopathy.resultsin.anirregular.structuredefectivechaperone-like.functionProc. Natl. Acad. Sci. U.S.A. 199996613742
  99. 99. TreweekT. M.RekasA.LindnerR. A.MJWalkerAquilina. J. A.RobinsonC. V.HorwitzJ.MDPerngQuinlan. R. A.CarverJ. A.R1alphaG.B-crystallinpromotes.theunfolding.ofreduced.alpha-lactalbuminisinherently.unstableFebs J. 200527271124
  100. 100. MDPerngCairns. L.van DenI.JsselP.PrescottA.HutchesonA. M.QuinlanR. A.Intermediate filament interactions can be altered by HSP27 and alphaB-crystallin. J Cell Sci 1999Pt 13):2099-112.
  101. 101. MDPerngWen. S. F.van denI.JsselP.PrescottA. R.QuinlanR. A.Desmin aggregate formation by R120G alphaB-crystallin is caused by altered filament interactions and is dependent upon network status in cells. Mol Biol Cell 200415233546
  102. 102. BerryV.FrancisP.MAReddyCollyer. D.VithanaE.MacKay. I.DawsonG.CareyA. H.MooreA.BhattacharyaS. S.QuinlanR. A.Alpha-Bcrystallin.gene. C. R. Y. A. B.mutationcauses.dominantcongenital.posteriorpolar.cataractin.humansAm. J. Hum. Genet. 20016911415
  103. 103. LiuY.ZhangX.LuoL.WuM.ZengR.ChengG.HuB.LiuB.LiangJ. J.ShangF. A.novelalpha.B-crystallinmutation.associatedwith.autosomaldominant.congenitallamellar.cataractInvest. Ophthalmol. Vis. Sci. 200647106975
  104. 104. LiuM.KeT.WangZ.YangQ.ChangW.JiangF.TangZ.LiH.RenX.WangX.WangT.LiQ.YangJ.LiuJ.WangQ. K.Identification of a CRYAB mutation associated with autosomal dominant posterior polar cataract in a Chinese family. Invest. Ophthalmol. Vis. Sci. 20064734616
  105. 105. DeviR. R.YaoW.VijayalakshmiP.SergeevY. V.SundaresanP.HejtmancikJ. F.Crystallin gene mutations in Indian families with inherited pediatric cataract. Mol. Vis. 200814115770
  106. 106. Safieh LA, Khan AO, Alkuraya FS.Identification of a novel CRYAB mutation associated with autosomal recessive juvenile cataract in a Saudi family. Mol. Vis. 2009159804
  107. 107. ChenQ.MaYanJ.MothobiM.MELiuY.ZhengF. A.novelmutation.inC. R. Y. A. B.associatedwith.autosomaldominant.congenitalnuclear.cataractin. a.Chinesefamily.Mol. Vis. 200915135965
  108. 108. SelcenD.EngelA. G.Myofibrillar myopathy caused by novel dominant negative alpha B-crystallin mutations. Ann. Neurol. 20035480410
  109. 109. Del BigioM. R.ChudleyA. E.SarnatH. B.CampbellC.GoobieS.ChodirkerB. N.SelcenD.Infantile muscular dystrophy in Canadian aboriginals is an αB-crystallinopathy. Ann. Neurol. 20116986671
  110. 110. ForrestK. M. L.Al-SarrajS.SewryC.BukS.TanS. V.PittM.DurwardA.Mc DougallM.IrvingM.HannaM. G.MatthewsE.SarkozyA.HudsonJ.BarresiR.BushbyK.JungbluthH.WraigeE.Infantile onset myofibrillar myopathy due to recessive CRYAB mutations. Neuromuscul Disord 2011213740
  111. 111. ReilichP.SchoserB.SchrammN.KrauseS.SchesslJ.KressW.Müller-HöckerJ.WalterM. C.LochmüllerH.Thep.G1mutationS.ofthe.alpha-Bcrystallin.gene. C. R. Y. A. B.causeslate-onset.distalmyopathy.Neuromuscul Disord 2010202559
  112. 112. PilottoA.MarzilianoN.PasottiM.GrassoM.CostanteA. M.ArbustiniE.alphaalphaB-crystallin mutation in dilated cardiomyopathies: low prevalence in a consecutive series of 200 unrelated probands. Biochem Biophys Res Commun 200634611157
  113. 113. InagakiN.HayashiT.ArimuraT.KogaY.TakahashiM.ShibataH.TeraokaK.ChikamoriT.YamashinaA.KimuraA.Alpha B-crystallin mutation in dilated cardiomyopathy. Biochem Biophys Res Commun 200634237986
  114. 114. SacconiS.FéassonL.AntoineJ. C.PécheuxC.BernardR.CoboA. M.CasarinA.SalviatiL.DesnuelleC.UrtizbereaA. A.novelC. R. Y. A. B.mutationresulting.inmultisystemic.diseaseNeuromuscul Disord 2012226672
  115. 115. VicartP.CaronA.GuicheneyP.LiZ.PrévostM. C.FaureA.ChateauD.ChaponF.ToméF.DupretJ. M.PaulinD.FardeauM. A.missensemutation.inthe.alphaB-crystallin.chaperonegene.causesa.desmin-relatedmyopathy.Nat. Genet. 199820925
  116. 116. KumarL. V.RamakrishnaT.RaoC. M.Structural and functional consequences of the mutation of a conserved arginine residue in alphaA and alphaB crystallins. J Biol Chem 19992742413741
  117. 117. MDPerngMuchowski. P. J.van DenI.JsselP.WuG. J.HutchesonA. M.ClarkJ. I.QuinlanR. A.The cardiomyopathy and lens cataract mutation in alphaB-crystallin alters its protein structure, chaperone activity, and interaction with intermediate filaments in vitro. J Biol Chem 19992743323543
  118. 118. WangX.OsinskaH.KlevitskyR.GerdesA. M.NiemanM.LorenzJ.HewettT.RobbinsJ.Expression of R120G-alphaB-crystallin causes aberrant desmin and alphaB-crystallin aggregation and cardiomyopathy in mice. Circ. Res. 2001898491
  119. 119. SanbeA.OsinskaH.SaffitzJ. E.GlabeC. G.KayedR.MaloyanA.RobbinsJ.Desmin-related cardiomyopathy in transgenic mice: a cardiac amyloidosis. Proc. Natl. Acad. Sci. U.S.A. 2004101101326
  120. 120. MaloyanA.SanbeA.OsinskaH.WestfallM.RobinsonD.Imahashi-IK.MurphyE.RobbinsJ.Mitochondrial dysfunction and apoptosis underlie the pathogenic process in alpha-B-crystallin desmin-related cardiomyopathy. Circulation 2005112345161
  121. 121. Engelsmanden. J.GerritsD.de JongW. W.RobbinsJ.KatoK.BoelensW. C.Nuclear import of {alpha}B-crystallin is phosphorylation-dependent and hampered by hyperphosphorylation of the myopathy-related mutant R120G. J Biol Chem 20052803713948
  122. 122. AndleyU. P.HamiltonP. D.RaviN.WeihlC. C. A.knock-inmouse.modelfor.theR1.mutationG.ofα.B-crystallinrecapitulates.humanhereditary.myopathycataractsP.LoPLoS ONE 2011e17671.
  123. 123. SimonS.MichielM.Skouri-PanetF.LechaireJ. P.VicartP.TardieuA.ResidueR1.isessential.forthe.quaternarystructure.functionalintegrity.ofhuman.alphaB-crystallin.Biochemistry 200746960514
  124. 124. HayesV. H.DevlinG.QuinlanR. A.Truncation of alphaB-crystallin by the myopathy-causing Q151X mutation significantly destabilizes the protein leading to aggregate formation in transfected cells. J Biol Chem 20082831050012
  125. 125. KohT. J.EscobedoJ.Cytoskeletal disruption and small heat shock protein translocation immediately after lengthening contractions. Am. J. Physiol., Cell Physiol. 2004C71322
  126. 126. KamradtM. C.ChenF.SamS.CrynsV. L.The small heat shock protein alpha B-crystallin negatively regulates apoptosis during myogenic differentiation by inhibiting caspase-3 activation. J Biol Chem 2002277387316
  127. 127. Li-CD. W.Liu-PJ.Mao-WY.XiangH.WangJ.Ma-YW.DongZ.PikeH. M.BrownR. E.ReedJ. C.Calcium-activatedR. A. F. M. E. K. E. R. K.signalingpathway.mediatesp53-dependent.apoptosisisabrogated.byalpha.B-crystallinthrough.inhibitionof. R. A. S.activationMol Biol Cell 200516443753
  128. 128. Adhikari AS, Singh BN, Rao KS, Rao CM. αB-crystallin, a small heat shock protein, modulates NF-κB activity in a phosphorylation-dependent manner and protects muscle myoblasts from TNF-α induced cytotoxicity.Biochim. Biophys. Acta 20111813153242
  129. 129. Brady JP, Garland DL, Green DE, Tamm ER, Giblin FJ, Wawrousek EF.AlphaB-crystallin in lens development and muscle integrity: a gene knockout approach. Invest. Ophthalmol. Vis. Sci. 200142292434
  130. 130. MoyanoJ. V.EvansJ. R.ChenF.LuM.MEWernerYehiely. F.DiazL. K.TurbinD.KaracaG.WileyE.NielsenT. O.PerouC. M.CrynsV. L.AlphaAlphaB-crystallin is a novel oncoprotein that predicts poor clinical outcome in breast cancer. J. Clin. Invest. 200611626170
  131. 131. GoplenD.BougnaudS.RajcevicU.BøeS. O.SkaftnesmoK. O.VogesJ.EngerP. Ø.WangJ.TysnesB. B.LaerumO. D.NiclouS.BjerkvigR. α.B-crystallin is elevated in highly infiltrative apoptosis-resistant glioblastoma cells. Am J Pathol 2010177161828
  132. 132. ChiesiM.LongoniS.LimbrunoU.Cardiacalpha-crystallin. I. I. I.Involvement during heart ischemia. Mol. Cell. Biochem. 19909712936
  133. 133. van NoortJ. M.van SechelA. C.BajramovicJ. J.Ouagmiri elM.PolmanC. H.LassmannH.RavidR.The small heat-shock protein alpha B-crystallin as candidate autoantigen in multiple sclerosis. Nature 1995375798801
  134. 134. RenkawekK.CEVoorterBosman. G. J.van WorkumF. P.de JongW. W.Expression of alpha B-crystallin in Alzheimer’s disease. Acta Neuropathol. 19948715560
  135. 135. ChávezZobel. A. T.LorangerA.MarceauN.ThériaultJ. R.LambertH.LandryJ.Distinct chaperone mechanisms can delay the formation of aggresomes by the myopathy-causing R120G alphaB-crystallin mutant. Hum. Mol. Genet. 200312160920
  136. 136. HishiyaA.SalmanM. N.CarraS.KampingaH. H.TakayamaS. B. A. G.directlyinteracts.withmutated.alphaB-crystallin.tosuppress.itsaggregation.toxicityP.LoPLoS ONE 2011e16828.
  137. 137. ItoH.KameiK.IwamotoI.InagumaY.KatoK.Regulation of the levels of small heat-shock proteins during differentiation of C2C12 cells. Exp Cell Res 200126621321
  138. 138. SugiyamaY.SuzukiA.KishikawaM.AkutsuR.HiroseT.MMWayeTsui. S. K.YoshidaS.OhnoS.Muscle develops a specific form of small heat shock protein complex composed of MKBP/HSPB2 and HSPB3 during myogenic differentiation. J Biol Chem 20002751095104
  139. 139. Singh BN, Rao KS, Rao CM.Ubiquitin-proteasome-mediated degradation and synthesis of MyoD is modulated by alphaB-crystallin, a small heat shock protein, during muscle differentiation. Biochim. Biophys. Acta 2010180328899
  140. 140. ReilichP.KrauseS.SchrammN.KlutznyU.BulstS.ZehetmayerB.SchneideratP.WalterM. C.SchoserB.LochmüllerH. A.novelmutation.inthe.myotilingene. . M. Y. O. T.causesa.severeform.oflimb.girdlemuscular.dystrophy. A. . L. G. M. D. A.J. Neurol. 2011258143744
  141. 141. MAHauserConde. C. B.KowaljowV.ZeppaG.TaratutoA. L.TorianU. M.VanceJ.MAPericak-VanceSpeer. M. C.RosaA. L.myotilin Mutation found in second pedigree with LGMD1A. Am. J. Hum. Genet. 200271142832
  142. 142. ShalabyS.MitsuhashiH.MatsudaC.MinamiN.NoguchiS.NonakaI.NishinoI.HayashiY. K.Defective myotilin homodimerization caused by a novel mutation in MYOT exon 9 in the first Japanese limb girdle muscular dystrophy 1A patient. J. Neuropathol. Exp. Neurol. 2009687017
  143. 143. SelcenD.EngelA. G.Mutations in myotilin cause myofibrillar myopathy. Neurology 200462136371
  144. 144. ForoudT. A.mutationin.myotilincauses.spheroidbody.myopathyNeurology 200565193640
  145. 145. Mc NeillA.BirchallD.StraubV.GoldfarbL.ReilichP.WalterM. C.SchrammN.Lochmuuml.llerH.ChinneryP. F.Lower Limb Radiology of Distal Myopathy due to the S60F Myotilin Mutation. Eur Neurol 2009621616
  146. 146. MykkänenO. M.GrönholmM.RöntyM.LalowskiM.SalmikangasP.SuilaH.CarpénO.Characterization of human palladin, a microfilament-associated protein. Mol Biol Cell 200112306073
  147. 147. Parast MM, Otey CA. Characterization of palladin, a novel protein localized to stress fibers and cell adhesions.J. Cell Biol. 200015064356
  148. 148. OlivéM.GoldfarbL. G.ShatunovA.FischerD.FerrerI.Myotilinopathyrefining.theclinical.myopathologicalphenotype.Brain 2005128231526
  149. 149. BercianoJ.GallardoE.Domínguez-PerlesR.GallardoE.GarcíaA.García-BarredoR.CombarrosO.InfanteJ.IllaI.Autosomal-dominant distal myopathy with a myotilin S55F mutation: sorting out the phenotype. J. Neurol. Neurosurg. Psychiatr. 2008792058
  150. 150. GamezJ.ArmstrongJ.ShatunovA.Selva-O’CallaghanA.Dominguez-OronozR.OrtegaA.GoldfarbL.FerrerI.OlivéM.Generalized muscle pseudo-hypertrophy and stiffness associated with the myotilin Ser55Phe mutation: a novel myotilinopathy phenotype? J. Neurol. Sci. 200927716771
  151. 151. Garvey SM, Miller SE, Claflin DR, Faulkner JA, Hauser MA.Transgenic mice expressing the myotilin T57I mutation unite the pathology associated with LGMD1A and MFM. Hum. Mol. Genet. 200615234862
  152. 152. Pénisson-BesnierI.TalvinenK.DumezC.ViholaA.DubasF.FardeauM.HackmanP.CarpénO.UddB.Myotilinopathy in a family with late onset myopathy. Neuromuscul Disord 20061642731
  153. 153. SalmikangasP.MykkänenO. M.GrönholmM.HeiskaL.KereJ.CarpénO.Myotilina.novelsarcomeric.proteinwith.twoIg-like.domainsis.encodedby. a.candidategene.forlimb-girdle.musculardystrophy.Hum. Mol. Genet. 19998132936
  154. 154. MologniL.MozaM.MMLalowskiCarpén. O.Characterization of mouse myotilin and its promoter. Biochem Biophys Res Commun 200532910019
  155. 155. SchroderR.ReimannJ.SalmikangasP.ClemenC. S.HayashiY. K.NonakaI.ArahataK.CarpénO.BeyondL. G. M. D. A.myotilinis. a.componentof.centralcore.lesionsnemalinerods.Neuromuscul Disord 2003134515
  156. 156. WangJ.DubeD. K.MittalB.SangerJ. M.SangerJ. W.Myotilin dynamics in cardiac and skeletal muscle cells. Cytoskeleton (Hoboken) 20116866170
  157. 157. GontierY.TaivainenA.FontaoL.SonnenbergA.van der FlierA.CarpénO.FaulknerG.BorradoriL.The Z-disc proteins myotilin and FATZ-1 interact with each other and are connected to the sarcolemma via muscle-specific filamins. J Cell Sci 2005118373949
  158. 158. Nandelstadhvon. P.IsmailM.GardinC.SuilaH.ZaraI.BelgranoA.ValleG.CarpénO.FaulknerG. A.classI. I. I. P. D. Z.bindingmotif.inthe.myotilinfamiliesF. A. T. Z.bindsenigma.familyproteins. a.commonlink.forZ-disc.myopathiesMol. Cell. Biol. 20092982234
  159. 159. van der VenP. F.WiesnerS.SalmikangasP.AuerbachD.HimmelM.KempaS.HayessK.PacholskyD.TaivainenA.SchroderR.CarpénO.DOFurstIndications for a novel muscular dystrophy pathway. gamma-filamin, the muscle-specific filamin isoform, interacts with myotilin. J. Cell Biol. 200015123548
  160. 160. WittS. H.GranzierH.WittC. C.LabeitS. M. U. R. F.M. U. R. F.Targeta.SpecificSubset.ofMyofibrillar.ProteinsRedundantly.TowardsUnderstanding. M. U. R.F-dependentMuscle.UbiquitinationJ. Mol. Biol. 200535071322
  161. 161. SalmikangasP.van der VenP. F. M.LalowskiM.TaivainenA.ZhaoF.SuilaH.SchröderR.LappalainenP.DOFürstCarpén. O.Myotilin, the limb-girdle muscular dystrophy 1A (LGMD1A) protein, cross-links actin filaments and controls sarcomere assembly. Hum. Mol. Genet. 200312189203
  162. 162. Nandelstadhvon. P.GrönholmM.MozaM.LambergA.SavilahtiH.CarpénO.Actin-organising properties of the muscular dystrophy protein myotilin. Exp Cell Res 20053101319
  163. 163. MozaM.MologniL.TrokovicR.FaulknerG.PartanenJ.CarpénO.Targeted deletion of the muscular dystrophy gene myotilin does not perturb muscle structure or function in mice. Mol. Cell. Biol. 20072724452
  164. 164. OchalaJ.CarpénO.LarssonL.Maintenance of muscle mass, fiber size, and contractile function in mice lacking the Z-disc protein myotilin. Ups. J. Med. Sci. 200911423541
  165. 165. BarrachinaM.MorenoJ.JuvésS.MorenoD.OlivéM.FerrerI.Target genes of neuron-restrictive silencer factor are abnormally up-regulated in human myotilinopathy. Am J Pathol 2007171131223
  166. 166. JanuéA.OlivéM.FerrerI.Oxidative stress in desminopathies and myotilinopathies: a link between oxidative damage and abnormal protein aggregation. Brain Pathol. 20071737788
  167. 167. OliveM.van LeeuwenF. W.JanuéA.MorenoD.Torrejón-EscribanoB.FerrerI.Expression of mutant ubiquitin (UBB +1) and 62in myotilinopathies and desminopathies. Neuropathol Appl Neurobiol 2007
  168. 168. GollD. E.NetiG.MaresS. W.ThompsonV. F.Myofibrillarprotein.turnoverThe.proteasomethecalpains.Journal of Animal Science 2007E19E35.
  169. 169. Nandelstadhvon. P.SoliymaniR.BaumannM.CarpénO.Analysis of myotilin turnover provides mechanistic insight into the role of myotilinopathy-causing mutations. Biochem. J. 201143611321
  170. 170. FaulknerG.PallaviciniA.FormentinE.ComelliA.IevolellaC.TrevisanS.BortolettoG.ScannapiecoP.SalamonM.MoulyV.ValleG.LanfranchiG. Z. A. S. P. a.newZ-band.alternativelyspliced. P. D.Z-motifprotein.J. Cell Biol. 199914646575
  171. 171. ZhouQ.Ruiz-LozanoP.MEMartoneChen. J.Cyphera.striatedmuscle-restricted. P. D. Z.domain-containingL. I. M.proteinbinds.toalpha-actinin.proteinkinase. C.J. Cypher, a striated muscle-restricted PDZ and LIM domain-containing protein, binds to alpha-actinin-2 and protein kinase C. J Biol Chem 19992741980713
  172. 172. PassierR.RichardsonJ. A.OlsonE. N.Oraclea.novel-LP. D. Z.domainI. M.proteinexpressed.inheart.skeletalmuscle.Mech Develop 20009227784
  173. 173. Guy PM, Kenny DA, Gill GN. The PDZ domain of the LIM protein enigma binds to beta-tropomyosin.Mol Biol Cell 199910197384
  174. 174. PomièsP.MacalmaT.BeckerleM. C.Purification and characterization of an alpha-actinin-binding PDZ-LIM protein that is up-regulated during muscle differentiation. J Biol Chem 19992742924250
  175. 175. ArimuraT.HayashiT.TeradaH.Lee-YS.ZhouQ.TakahashiM.UedaK.NouchiT.HohdaS.ShibutaniM.HiroseM.ChenJ.Park-EJ.YasunamiM.HayashiH.KimuraA. A.CypherZ. A. S. P.mutationassociated.withdilated.cardiomyopathyalters.thebinding.affinityto.proteinkinase. C.J, Park J-E, Yasunami M, Hayashi H, Kimura A. A Cypher/ZASP mutation associated with dilated cardiomyopathy alters the binding affinity to protein kinase C. J Biol Chem 2004279674652
  176. 176. TheisJ. L.BosJ. M.BartlesonV. B.WillM. L.BinderJ.VattaM.TowbinJ. A.GershB. J.OmmenS. R.MJAckermanEchocardiographic-determined septal morphology in Z-disc hypertrophic cardiomyopathy. Biochem Biophys Res Commun 2006351896902
  177. 177. SelcenD.EngelA. G.Mutationsin. Z. A. S. P.definea.novelform.ofmuscular.dystrophyin.humansAnn. Neurol. 20055726976
  178. 178. CaiH.YabeI.SatoK.KanoT.NakamuraM.HozenH.SasakiH.Clinicalpathological.geneticmutation.analysisof.sporadicinclusion.bodymyositis.inJapanese.peopleJ. Neurol. 2012
  179. 179. AuY.AtkinsonR. A.GuerriniR.KellyG.JosephC.MartinS. R.MuskettF. W.PallaviciniA.FaulknerG.PastoreA.Solution structure of ZASP PDZ domain; implications for sarcomere ultrastructure and enigma family redundancy. Structure 20041261122
  180. 180. KlaavuniemiT.KelloniemiA.YlänneJ.The ZASP-like motif in actinin-associated LIM protein is required for interaction with the alpha-actinin rod and for targeting to the muscle Z-line. J Biol Chem 20042792640210
  181. 181. KlaavuniemiT.YlanneJ.ZaspZasp/Cypher internal ZM-motif containing fragments are sufficient to co-localize with alpha-actinin- Analysis of patient mutations. Exp Cell Res 20063121299311
  182. 182. FreyN.OlsonE.Calsarcin-novela.skeletalmuscle-specific.memberof.thecalsarcin.familyinteracts.withmultiple.Z-discproteins.J Biol Chem 2002277139984004
  183. 183. McKeown CR, Han H-F, Beckerle MC.Molecular characterization of the Caenorhabditis elegans ALP/Enigma gene alp-1. Dev Dyn 20062355308
  184. 184. JaniK.SchöckF.Zasp is required for the assembly of functional integrin adhesion sites. J. Cell Biol. 2007179158397
  185. 185. BennaC.PeronS.RizzoG.FaulknerG.MegighianA.PeriniG.TognonG.ValleG.ReggianiC.CostaR.MAZordanPost-transcriptional silencing of the Drosophila homolog of human ZASP: a molecular and functional analysis. Cell Tissue Res. 200933746376
  186. 186. KatzemichA.LongJ. Y.JaniK.LeeB. R.SchöckF.Muscle type-specific expression of Zasp52 isoforms in Drosophila. Gene Expr. Patterns 20111148490
  187. 187. van der MeerD. L. M.MarquesI. J.LeitoJ. T. D.BesserJ.BakkersJ.SchoonheereE.CPBagowskiZebrafish cypher is important for somite formation and heart development. Dev. Biol. 200629935672
  188. 188. HuangC.ZhouQ.LiangP.MSHollanderSheikh. F.LiX.GreaserM.SheltonG. D.EvansS.ChenJ.Characterization and in vivo functional analysis of splice variants of cypher. J Biol Chem 200327873605
  189. 189. ChengH.ZhengM.PeterA. K.KimuraK.LiX.OuyangK.ShenT.CuiL.FrankD.DaltonN. D.GuY.FreyN.PetersonK. L.EvansS. M.KnowltonK. U.SheikhF.ChenJ.Selective deletion of long but not short Cypher isoforms leads to late-onset dilated cardiomyopathy. Hum. Mol. Genet. 201120175162
  190. 190. ZhouQ.ChuP. H.HuangC.ChengC. F.MEMartoneKnoll. G.SheltonG. D.EvansS.ChenJ.Ablationof.Cypher-La. P. D. Z.domainI. M.Z-lineprotein.causesa.severeform.ofcongenital.myopathyJ. Ablation of Cypher, a PDZ-LIM domain Z-line protein, causes a severe form of congenital myopathy. J. Cell Biol. 200115560512
  191. 191. XingY.IchidaF.MatsuokaT.IsobeT.IkemotoY.HigakiT.TsujiT.HanedaN.KuwabaraA.ChenR.FutataniT.TsubataS.WatanabeS.WatanabeK.HironoK.UeseK.MiyawakiT.BowlesK. R.BowlesN. E.TowbinJ. A.Genetic analysis in patients with left ventricular noncompaction and evidence for genetic heterogeneity. Mol. Genet. Metab. 200688717
  192. 192. GriggsR.ViholaA.HackmanP.TalvinenK.HaravuoriH.FaulknerG.EymardB.RichardI.SelcenD.EngelA.CarpénO.UddB.Zaspopathy in a large classic late-onset distal myopathy family. Brain 2007130147784
  193. 193. LiZ.AiT.SamaniK.XiY.Tzeng-PH.XieM.WuS.GeS.MDTaylor-WDong. J.ChengJ.MJAckermanKimura. A.SinagraG.BrunelliL.FaulknerG.VattaM. A. Z. A. S. P.missensemutation.S1leadsL.tocytoskeletal.electricalabnormalities.ina.mousemodel.ofcardiomyopathy.Circ Arrhythm Electrophysiol 2010364656
  194. 194. ZhengM.ChengH.LiX.ZhangJ.CuiL.OuyangK.HanL.ZhaoT.GuY.DaltonN. D.Bang-LM.PetersonK. L.ChenJ.Cardiac-specific ablation of Cypher leads to a severe form of dilated cardiomyopathy with premature death. Hum. Mol. Genet. 20091870113
  195. 195. Mochly-RosenD.WuG.HahnH.OsinskaH.LironT.LorenzJ. N.YataniA.RobbinsJ.DornG. W.Cardiotrophic effects of protein kinase C epsilon: analysis by in vivo modulation of PKCepsilon translocation. Circ. Res. 20008611739
  196. 196. PassJ. M.ZhengY.WeadW. B.ZhangJ.LiR. C.BolliR.PingP. P. K.PKCepsilon activation induces dichotomous cardiac phenotypes and modulates PKCepsilon-RACK interactions and RACK expression. Am. J. Physiol. Heart Circ. Physiol. 2001H94655
  197. 197. Johnson JA, Gray MO, Chen CH, Mochly-Rosen D. A protein kinase C translocation inhibitor as an isozyme-selective antagonist of cardiac function. J Biol Chem 1996;271:24962-6.
  198. 198. LiuG. S.CohenM. V.Mochly-RosenD.DowneyJ. M.Protein kinase C-epsilon is responsible for the protection of preconditioning in rabbit cardiomyocytes. J. Mol. Cell. Cardiol. 199931193748
  199. 199. WakasakiH.KoyaD.SchoenF. J.JirousekM. R.WaysD. K.BDHoitWalsh. R. A.KingG. L.Targeted overexpression of protein kinase C beta2 isoform in myocardium causes cardiomyopathy. Proc. Natl. Acad. Sci. U.S.A. 19979493205
  200. 200. GoldspinkP. H.MontgomeryD. E.WalkerL. A.UrbonieneD.Mc KinneyR. D.GeenenD. L.SolaroR. J.ButtrickP. M.Protein kinase Cepsilon overexpression alters myofilament properties and composition during the progression of heart failure. Circ. Res. 20049542432
  201. 201. TakeishiY.PingP.BolliR.KirkpatrickD. L.BDHoitWalsh. R. A.Transgenic overexpression of constitutively active protein kinase C epsilon causes concentric cardiac hypertrophy. Circ. Res. 200086121823
  202. 202. ArimuraT.InagakiN.HayashiT.ShichiD.SatoA.HinoharaK.VattaM.TowbinJ. A.ChikamoriT.YamashinaA.KimuraA.Impaired binding of ZASP/Cypher with phosphoglucomutase 1 is associated with dilated cardiomyopathy. Cardiovasc. Res. 200983808
  203. 203. ShatunovA.eacuteM. O.OdgerelZ.Stadelmann-NesslerC.IrlbacherK.van LandeghemF.BayarsaikhanM.Lee-SH.GoudeauB.ChinneryP. F.StraubV.Hilton-JonesD.MSDamianKaminska. A.VicartP.BushbyK.DalakasM. C.SambuughinN.FerrerI.GoebelH. H.GoldfarbL. G.In-frame deletion in the seventh immunoglobulin-like repeat of filamin C in a family with myofibrillar myopathy 20091765663
  204. 204. LuanX.HongD.ZhangW.WangZ.YuanY. A.novelheterozygous.deletion-insertionmutation.(2695-27del insG. T. T. T. G. T.inexon. .ofthe.filaminC.genecauses.filaminopathyin. a.largeChinese.familyNeuromuscul Disord 2010203906
  205. 205. DuffR. M.TayV.HackmanP.RavenscroftG.Mc LeanC.KennedyP.SteinbachA.SchöfflerW.van der VenP. F. M.DOFürstSong. J.Djinović-CarugoK.PenttiläS.RaheemO.ReardonK.MalandriniA.GambelliS.VillanovaM.NowakK. J.WilliamsD. R.LandersJ. E.BrownR. H.Jr UddB.LaingN. G.Mutations in the N-terminal Actin-Binding Domain of Filamin C Cause a Distal Myopathy. Am. J. Hum. Genet. 20112011112
  206. 206. GuergueltchevaV.PeetersK.BaetsJ.Ceuterick-deGroote. C.MartinJ. J.SulsA.De VriendtE.MihaylovaV.ChamovaT.Almeida-SouzaL.YdensE.TzekovC.HadjidekovG.GospodinovaM.StormK.ReyniersE.BichevS.van der VenP. F. M.DOFurstMitev. V.LochmullerH.TimmermanV.TournevI.De JongheP.JordanovaA.Distal myopathy with upper limb predominance caused by filamin C haploinsufficiency. Neurology 2011
  207. 207. LoweT.KleyR. A.van der VenP. F. M.HimmelM.HuebnerA.VorgerdM.DOFurstThe pathomechanism of filaminopathy: altered biochemical properties explain the cellular phenotype of a protein aggregation myopathy. Hum. Mol. Genet. 20071613518
  208. 208. MaestriniE.PatrossoC.ManciniM.RivellaS.RocchiM.RepettoM.VillaA.FrattiniA.ZoppèM.VezzoniP.Mapping of two genes encoding isoforms of the actin binding protein ABP-280, a dystrophin like protein, to Xq28 and to chromosome 7. Hum. Mol. Genet. 199327616
  209. 209. GorlinJ. B.HenskeE.WarrenS. T.KunstC. B. D.aposUrso. M.PalmieriG.HartwigJ. H.BrunsG.KwiatkowskiD. J.Actin-bindingprotein. . A. B.P-28filamingene. . F. L. N.mapstelomeric.tothe.colorvision.locus. R. G. C. P.centromericto. G. P. D.inXq2.Genomics 1993174968
  210. 210. XieZ.XuW.DavieE. W.ChungD. W.Molecular cloning of human ABPL, an actin-binding protein homologue. Biochem Biophys Res Commun 19982519149
  211. 211. TakafutaT.WuG. X.MurphyG. F.ShapiroS. S.Human beta-filamin is a new protein that interacts with the cytoplasmic tail of glycoprotein Ib alpha. J Biol Chem 1998273175318
  212. 212. XuW. F.XieZ. W.ChungD. W.DavieE. W. A.novelhuman.actin-bindingprotein.homologuethat.bindsto.plateletglycoprotein.Ibalpha.Blood 199892126876
  213. 213. ThompsonT. G.ChanY. M.AAHackBrosius. M.RajalaM.LidovH. G.Mc NallyE. M.WatkinsS.KunkelL. M.Filamin. . F. L.N2muscle-specificA.sarcoglycaninteracting.proteinJ. Cell Biol. 200014811526
  214. 214. BeathamJ.RomeroR.TownsendS. K. M.HackerT.van der VenP. F. M.BlancoG.Filamin C interacts with the muscular dystrophy KY protein and is abnormally distributed in mouse KY deficient muscle fibres. Hum. Mol. Genet. 200413286374
  215. 215. van der FlierA.SonnenbergA.Structural and functional aspects of filamins. Biochim. Biophys. Acta 2001153899117
  216. 216. HimmelM.van der VenP. F. M.StockleinW.DOFurstThe limits of promiscuity: Isoform-specific dimerization of filamins. Biochemistry 2003424309
  217. 217. van der VenP. F. M.EhlerE.VakeelP.EulitzS.SchenkJ. A.MiltingH.MicheelB.DOFürstUnusual splicing events result in distinct Xin isoforms that associate differentially with filamin c and Mena/VASP. Exp Cell Res 2006312215467
  218. 218. FaulknerG.PallaviciniA.ComelliA.SalamonM.BortolettoG.IevolellaC.TrevisanS.KojicS.DallaVecchia. F.LavederP.ValleG.LanfranchiG. F. A. T. Z. a.filaminactinintelethonin-bindingprotein.ofthe.Z-discof.skeletalmuscle.J Biol Chem 20002754123442
  219. 219. TakadaF.VanderWoude. D. L.TongH. Q.ThompsonT. G.WatkinsS. C.KunkelL. M.BeggsA. H.Myozeninan.alpha-actiningamma-filamin-bindingprotein.ofskeletal.muscleZ.linesProc. Natl. Acad. Sci. U.S.A. 2001981595600
  220. 220. ZhangM.LiuJ.ChengA.De YoungS. M.SaltielA. R.Identificationof. C. A. P.ascostamerica.proteinthat.interactswith.filaminC.Mol Biol Cell 200718473140
  221. 221. MaiweilidanY.KlauzaI.KordeliE.Novel interactions of ankyrins-G at the costameres: The muscle-specific Obscurin/Titin-Binding-related Domain (OTBD) binds plectin and filamin C. Exp Cell Res 201131772436
  222. 222. Holmes WB, Moncman CL.Nebulette interacts with filamin C. Cell Motil. Cytoskeleton 20086513042
  223. 223. DalkilicI.SchiendaJ.ThompsonT. G.KunkelL. M.Lossof.FilaminC. . F. L.NcResults.inSevere.Defectsin.MyogenesisMyotubeStructure.Mol. Cell. Biol. 200626652234
  224. 224. FujitaM.MitsuhashiH.IsogaiS.NakataT.KawakamiA.NonakaI.NoguchiS.HayashiY. K.NishinoI.KudoA.FilaminC.playsan.essentialrole.inthe.maintenanceof.thestructural.integrityof.cardiacskeletalmuscles.revealedby.themedaka.mutantzacro.Dev. Biol. 20112011111
  225. 225. NortonN.LiD.MJRiederSiegfried.JDRampersaudE.ZüchnerS.MangosS.Gonzalez-QuintanaJ.WangL.Mc GeeS.ReiserJ.MartinE.NickersonD. A.HershbergerR. E.Genome-wide Studies of Copy Number Variation and Exome Sequencing Identify Rare Variants in BAG3 as a Cause of Dilated Cardiomyopathy. Am. J. Hum. Genet. 20118827382
  226. 226. ArimuraT.IshikawaT.NunodaS.KawaiS.KimuraA.Dilatedcardiomyopathy-associated. B. A. G.mutationsimpair.Z-discassembly.enhancesensitivity.toapoptosis.incardiomyocytes.Hum. Mutat. 201132148191
  227. 227. LeeH.CherkS.ChanS.WongS.TongT.HoW.ChanA.LeeK.MakC. B. A.BAG3-related myofibrillar myopathy in a Chinese family. Clin. Genet. 2012813948
  228. 228. HommaS.IwasakiM.SheltonG. D.EngvallE.ReedJ. C.TakayamaS. B. A. G.deficiencyresults.infulminant.myopathyearlylethality.Am J Pathol 200616976173
  229. 229. RosatiA.GrazianoV.De LaurenziV.PascaleM.TurcoM. C. B. A. G.multifaceteda.proteinthat.regulatesmajor.cellpathways.Cell Death Dis 2011e141.
  230. 230. HishiyaA.KitazawaT.TakayamaS. B. A. G.Hscinteractwith.actincapping.proteinCap. Z.tomaintain.myofibrillarintegrity.undermechanical.stressCirc. Res. 2010107122031
  231. 231. WarrenG. L.SummanM.GaoX.ChapmanR.HuldermanT.SimeonovaP. P.Mechanisms of skeletal muscle injury and repair revealed by gene expression studies in mouse models. J. Physiol. (Lond.) 200758282541
  232. 232. SanoudouD.MACorbettHan. M.GhoddusiM.Nguyen-AM.VlahovichT.HardemanN.BeggsE. C.A. H.Skeletal muscle repair in a mouse model of nemaline myopathy. Hum. Mol. Genet. 200615260312
  233. 233. DoongH.PriceJ.KimY. S.GasbarreC.ProbstJ.LiottaL. A.BlanchetteJ.RizzoK.KohnE. C. A. I.R-B. A. G.formsan. E. G.F-regulatedternary.complexwith.phospholipaseC-gamma.Hsp7Hsc7Oncogene 200019438595
  234. 234. TakayamaS.ReedJ. C.Molecular chaperone targeting and regulation by BAG family proteins. Nat Cell Biol 2001E23741
  235. 235. DoongH.VrailasA.KohnE. C.What’sin.the’BA.G’?-afunctional.domainanalysis.ofthe. B. A. G.familyproteins.Cancer Letters 200220022532
  236. 236. CarraS.SeguinS. J.LambertH.LandryJ.HspB.chaperoneactivity.towardpoly.Q)-containingproteins.dependson.itsassociation.withBag.stimulatora.ofmacroautophagy.J, Lambert H, Landry J. HspB8 chaperone activity toward poly(Q)-containing proteins depends on its association with Bag3, a stimulator of macroautophagy. J Biol Chem 2008283143744
  237. 237. TakayamaS.XieZ.ReedJ. C.An evolutionarily conserved family of Hsp70/Hsc70 molecular chaperone regulators. J Biol Chem 19992747816
  238. 238. Mc ClellanA. J.FrydmanJ.Molecular chaperones and the art of recognizing a lost cause. Nat Cell Biol 2001E513
  239. 239. LeeJ. H.TakahashiT.YasuharaN.InazawaJ.KamadaS.TsujimotoY.Bisa.Bcl-2-bindingprotein.thatsynergizes.withBcl.inpreventing.celldeath.Oncogene 199918618390
  240. 240. OdgerelZ.SarkozyA.Lee-SH.Mc KennaC.RankinJ.StraubV.LochmüllerH.PaolaF.D’AmicoA.BertiniE.BushbyK.GoldfarbL. G.Inheritance patterns and phenotypic features of myofibrillar myopathy associated with a BAG3 mutation. Neuromuscul Disord 20102043842
  241. 241. FuchsM.PoirierD. J.SeguinS. J.LambertH.CarraS.CharetteS. J.LandryJ.Identification of the key structural motifs involved in HspB8/HspB6-Bag3 interaction. Biochem. J. 201042524555
  242. 242. JDLünemannSchmidt. J.SchmidD.BarthelK.WredeA.DalakasM. C.MünzC.Beta-amyloid is a substrate of autophagy in sporadic inclusion body myositis. Ann. Neurol. 20076147683
  243. 243. AntokuK.MaserR. S.ScullyW. J.DelachS. M.JohnsonD. E.Isolationof.Bclbindingproteins.thatexhibit.homologywith. B. A. G.suppressorof.deathdomains.proteinBiochem Biophys Res Commun 2001286100310
  244. 244. Bonelli P, Petrella A, Rosati A, Romano MF, Lerose R, Pagliuca MG, Amelio T, Festa M, Martire G, Venuta S, Turco MC, Leone A. BAG3 protein regulates stress-induced apoptosis in normal and neoplastic leukocytes. Leukemia 2004;18:358-60.
  245. 245. LiaoQ.OzawaF.FriessH.ZimmermannA.TakayamaS.ReedJ. C.KleeffJ.BüchlerM. W.The anti-apoptotic protein BAG-3 is overexpressed in pancreatic cancer and induced by heat stress in pancreatic cancer cell lines. FEBS Lett. 20015031517
  246. 246. PagliucaM. G.LeroseR.CiglianoS.LeoneA.Regulation by heavy metals and temperature of the human BAG-3 gene, a modulator of Hsp70 activity. FEBS Lett. 2003541115
  247. 247. ChenL.WuW.DentchevT.ZengY.WangJ.TsuiI.TobiasJ. W.BennettJ.BaldwinD.DunaiefJ. L.Light damage induced changes in mouse retinal gene expression. Exp. Eye Res. 20047923947
  248. 248. RomanoM. F.FestaM.PagliucaG.LeroseR.BisogniR.ChiurazziF.StortiG.VolpeS.VenutaS.TurcoM. C.LeoneA. B. A. G.proteincontrols.B-chroniclymphocytic.leukaemiacell.apoptosisCell Death Differ. 2003103835
  249. 249. SetoJ. T.LekM.QuinlanK. G. R.HouwelingP. J.ZhengX. F.GartonF.MacArthur. D. G.RafteryJ. M.GarveyS. M.MAHauserYang. N.HeadS. I.NorthK. N.Deficiencyof.α-actininisassociated.withincreased.susceptibilityto.contraction-induceddamage.skeletalmuscle.remodelingHum. Mol. Genet. 201120291427

Written By

Avnika Ruparelia, Raquel Vaz and Robert Bryson-Richardson

Submitted: 23 November 2011 Published: 22 August 2012