2044-5040-1-30 2044-5040 Review <p>ColVI myopathies: where do we stand, where do we go?</p> AllamandValériev.allamand@institut-myologie.org BriñasLaural.brinas@institut-myologie.org RichardPascalepascale.richard@psl.aphp.fr StojkovicTanyatanya.stojkovic@psl.aphp.fr Quijano-RoySusanasusana.quijano-roy@rpc.aphp.fr BonneGisèleg.bonne@institut-myologie.org

Inserm, U974, Paris, France

CNRS, UMR7215, Paris, France

UPMC Univ Paris 06 UM76, IFR14, Paris, France

Institut de Myologie, Paris, France

AP-HP, Groupe Hospitalier Pitié-Salpêtrière, UF Cardiogénétique et Myogénétique, Service de Biochimie Métabolique, Paris, France

Centre de Référence Neuromusculaire Paris-Est, Institut de Myologie, Groupe Hospitalier Pitié-Salpêtrière, Paris, France

AP-HP, Service de Pédiatrie, Centre de Référence Maladies Neuromusculaires (GNMH) Hôpital Raymond Poincaré, Garches, France

Skeletal Muscle 2044-5040 2011 1 1 30 http://www.skeletalmusclejournal.com/content/1/1/30 2194339110.1186/2044-5040-1-30
2432011239201123920112011Allamand et al; licensee BioMed Central Ltd.This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Collagen VI myopathies, caused by mutations in the genes encoding collagen type VI (ColVI), represent a clinical continuum with Ullrich congenital muscular dystrophy (UCMD) and Bethlem myopathy (BM) at each end of the spectrum, and less well-defined intermediate phenotypes in between. ColVI myopathies also share common features with other disorders associated with prominent muscle contractures, making differential diagnosis difficult. This group of disorders, under-recognized for a long time, has aroused much interest over the past decade, with important advances made in understanding its molecular pathogenesis. Indeed, numerous mutations have now been reported in the COL6A1, COL6A2 and COL6A3 genes, a large proportion of which are de novo and exert dominant-negative effects. Genotype-phenotype correlations have also started to emerge, which reflect the various pathogenic mechanisms at play in these disorders: dominant de novo exon splicing that enables the synthesis and secretion of mutant tetramers and homozygous nonsense mutations that lead to premature termination of translation and complete loss of function are associated with early-onset, severe phenotypes. In this review, we present the current state of diagnosis and research in the field of ColVI myopathies. The past decade has provided significant advances, with the identification of altered cellular functions in animal models of ColVI myopathies and in patient samples. In particular, mitochondrial dysfunction and a defect in the autophagic clearance system of skeletal muscle have recently been reported, thereby opening potential therapeutic avenues.

Review

Collagen VI: an important component of connective tissues

Collagens are major constituents of the extracellular matrix (ECM), and are found in most connective tissues. They provide structural and mechanical stability to tissues, but they also play crucial roles in cell-ECM interactions through various receptors 1. In particular, collagen type VI (ColVI), an important component of skeletal muscle ECM, is involved in maintaining tissue integrity by providing a structural link between different constituents of connective-tissue basement membranes (for example, collagen types I and IV, biglycan, and decorin) and cells 23456789101112131415 (Figure 1). In addition to its structural role, ColVI supports adhesion, spreading and migration of cells, and cell survival, as discussed later in this review.

<p>Figure 1</p>

Schematic representation of the collagen type VI (ColVI) intracellular assembly process, and interactions with skeletal muscle extracellular matrix (ECM) components

Schematic representation of the collagen type VI (ColVI) intracellular assembly process, and interactions with skeletal muscle extracellular matrix (ECM) components. Individual α(VI) chains fold through their triple helical domains to form monomers (1:1:1 ratio) in the endoplasmic reticulum (ER), which further align in an anti-parallel manner as dimers and tetramers that are stabilized by disulfide bonds between cysteine residues (S = S links). Post-translational modifications (indicated in orange) take place in the ER and Golgi, followed by secretion of tetramers that align non-covalently end to end, to form beaded microfibrils in the ECM. ColVI interacts with collagenous and non-collagenous components of the basal lamina and interstitial matrix surrounding muscle fibers.

ColVI is a heterotrimeric molecule composed of three individual α(VI) chains that display a similar structure, with a triple helical domain characterized by the repetition of the Gly-X-Y amino acid sequence, flanked by globular domains homologous to von Willebrand factor A domains 1617. In addition to the well-known α1(VI), α2(VI) and α3(VI) chains encoded in human by the COL6A1, COL6A2 (located head-to-tail on chromosome 21q22.3), and COL6A3 (on chromosome 2q37) genes 18, three novel chains, α4(VI), α5(VI) and α6(VI), have recently been identified 1920. These chains have high structural homology to the α3(VI) chain. In humans, the COL6A4, COL6A5 and COL6A6 genes are all located on chromosome 3q22.1, with the COL6A4 gene being split by a chromosome break and thus not coding for a protein 192021. The murine orthologs of these genes are organized in tandem on chromosome 9 (Col6a4, Col6a5 and Col6a6) and encode the α4(VI), α5(VI) and α6(VI) chains. The expression pattern of the three novel chains differs between mice and humans, and also between fetal and adult tissues 1920. Importantly, in the context of ColVI myopathies, the α6(VI) chain is the only one expressed at high levels in human skeletal muscle, at higher levels in fetal than adult tissue 19. In skin, a detailed analysis of the expression of the human α5(VI) and α6(VI) chains revealed that both chains are expressed, albeit differently, and that they are variably altered in tissues from patients with mutations in the COL6A1, COL6A2 and COL6A3 genes 22. Interestingly, the COL6A5 gene had previously been reported as associated with atopic dermatitis under the name COL29A1 23, but this association has recently been questioned 2425. The knee osteoarthritis susceptibility locus DVWA was shown to correspond to the 5' part of the split COL6A4 gene 21.

Although largely ubiquitous, the expression of ColVI seems to be finely regulated in different cell types and tissues, as shown for the murine Col6a1 gene. The identification of a transcriptional enhancer located in the 5'-flanking sequence of the gene points to a collaborative crosstalk between myogenic and mesenchymal/endomysial cells, enabling transcription of ColV in muscle connective tissue 262728.

The α1(VI), α2(VI) and α3(VI) chains assemble intracellularly as monomers (1:1:1 ratio), from their C-terminal ends, and subsequently form dimers (two anti-parallel, overlapping monomers) and tetramers (four monomers) that are stabilized by disulfide bonds between cysteine residues of the three chains 293031323334. ColVI chains are subjected to extensive post-translational modifications such as hydroxylation of lysine and proline residues 35, and glycosylation of hydroxylysines, which have been shown to be essential for the tetramerization and further secretion of ColVI 3637. Upon secretion, tetramers are further aligned end to end as microfibrils in the extracellular space, with a characteristic beaded appearance 33 (Figure 1). To date, somewhat contradictory results have been obtained regarding the possible assembly of the newly characterized α(VI) polypeptides with the α1(VI), α2(VI) chains. In transfection experiments, only α4(VI) appeared to have this ability 19, whereas in mouse muscle, all three were reported to do so 20. Whether and how these additional chains may fit in the pathogenesis of ColVI myopathies remains unresolved to date, and needs to be addressed more comprehensively. To date, in our cohort of patients, no pathogenic mutations have been found by sequencing of the COL6A5 and COL6A6 genes in patients without mutations in the COL6A1-3 genes (V. Allamand, data not shown).

Clinical phenotypes of collagen VI myopathies

The etiological definition of ColVI myopathies as a specific condition has evolved over the years with the blurring of boundaries between two disorders, initially described separately but now recognized as the extreme ends of a continuous clinical spectrum 3839 (Figure 2). The severe endpoint of this spectrum corresponds to Ullrich congenital muscular dystrophy (UCMD, OMIM 254090; http://www.ncbi.nlm.nih.gov/omim), described in 1930 as 'congenital atonic-sclerotic muscular dystrophy', emphasizing its early onset and the presence of proximal joint contractures associated with a striking distal hyperlaxity 4041. Orthopedic deformities (joint contractures, scoliosis) and respiratory impairment with diaphragmatic failure generally develop within the first decade of life, and may be life-threatening. Arrest of motor milestones with no acquisition of walking ability is seen in a subset of patients, but most children are able to walk, and show later progression of muscle weakness with loss of ambulation around 10 years of age, and a requirement for mechanical ventilation in late childhood or young adulthood 4243.

<p>Figure 2</p>

Clinical spectrum, associated spine deformation and muscle MRI in collagen type VI (ColVI) myopathies

Clinical spectrum, associated spine deformation and muscle MRI in collagen type VI (ColVI) myopathies. (A), Early severe phenotypes, corresponding to classic Ullrich congenital muscular dystrophy (UCMD), (B) intermediate forms seen in children or adults and (C) less severe, classic Bethlem myopathy (BM) forms constitute the overlapping clinical presentations of ColVI myopathies. (D) Radiography showing the evolution of spine deformation in a patient presenting with a classic early-onset UCMD phenotype. T1 transverse section of Bethlem myopathy upper limb girdle (E) and (F) thighs. Note the fatty infiltration, which appears as hyperintense area on T1-weighted images, located around the triceps brachialis muscles in (E) and along the fascia of vastus lateralis and vastus medialis muscles in (F) (yellow arrows). (G) The concentric fatty involvement of the thigh muscles is also seen on whole body MRI. (Images courtesy of Drs Susana Quijano-Roy and Tanya Stojkovic).

At the other end of the spectrum is the milder form Bethlem myopathy (BM, OMIM 158810), described in 1976, which begins in the first or second decade, although a neonatal history may be recognized, characterized by early contractures of finger flexors, wrist, elbows and ankles 4445. Respiratory failure and distal hyperlaxity are usually absent or are milder than in UCMD, although the latter may not be so uncommon in very young children with BM. The course is usually slow, with most of the patients remaining ambulatory. However, progression of muscle weakness occurs often in the fifth decade, resulting in about 50% of patients requiring walking aids or a wheelchair 46. Intermediate phenotypes have been described, and named 'mild UCMD' or 'severe BM', thereby reinforcing the notion of clinical overlap between Ullrich and Bethlem phenotypes 3839.

Skin features such as follicular hyperkeratosis and hypertrophic scars or keloid formation are common 38394243474849. Other common findings include normal cognitive abilities, normal or only slightly raised serum creatine kinase (CK) levels, and absence of cardiac phenotype. Two other conditions that fall within the spectrum of ColVI myopathies have been documented: autosomal dominant limb-girdle muscular dystrophy (LGMD) (in three families) and, more recently, autosomal recessive myosclerosis myopathy (OMIM 255600) (in one family) 5051.

The prevalences of UCMD and BM in northern England has recently been reported as 0.13 and 0.77 per 100,000, respectively, amounting collectively to 0.9 per 100,000 52. UCMD seems to be the second most common type of congenital muscular dystrophy (CMD) in Europe (behind laminin α2 chain deficiency; OMIM 607855) and also in Japan (behind Fukuyama congenital muscular dystrophy; OMIM 253800, 53) and Australia (behind α-dystroglycan glycosylation defects; 54). In the cohort from northern England, BM emerges as the fourth most common myopathy behind myotonic dystrophy (OMIM 160900), facio-scapulo-humeral muscular dystrophy (OMIM 158900) and Duchenne/Becker muscular dystrophy (OMIM 310200 and 300376) 52.

Differential diagnosis of ColVI-related myopathies

With the most prominent clinical presentation of ColVI myopathies being muscle weakness and contractures, associated with variable degrees of hyperlaxity, an important difficulty lies in defining boundaries and contiguities, with the possible differential diagnosis including congenital myopathies, Emery-Dreifuss muscular dystrophy (EDMD; OMIM 181350), LGMD, rigid spine muscular dystrophies, and other diseases of connective tissues such as Ehlers-Danlos syndrome 555657. Imaging techniques, such as computed tomography or magnetic resonance imaging (MRI) of muscle, are now recognized as very helpful in the diagnostic approach of muscle disease, because there are specific patterns of muscle involvement in each of these contractile myopathies as reported for EDMD with LMNA mutations 58, muscular dystrophies with rigidity of the spine 59, and ColVI myopathies 586061. From these studies, the typical pattern of muscle involvement in ColVI myopathies is now considered to be constituted by a diffuse, concentric hypodensity of the thigh muscles with relative sparing of the sartorius, gracilis and adductor longus muscles. The vasti muscles are the most affected muscles. In addition, a peculiar central area of abnormal signal is seen within the rectus femoris, initially referred to as a 'central shadow' 62.

In the context of the differential diagnosis, the absence of raised CK levels, the lack of a cardiac phenotype, and the presence of a specific MRI pattern are strongly suggestive of a ColVI myopathy.

Molecular diagnosis and genetics

In light of the clinical variability and the overlapping presentation with other muscular disorders, a definite diagnosis can only be made after the identification of pathogenic mutations in one of the COL6A genes, which to date are restricted to COL6A1, COL6A2 and COL6A3. However, the large size (106 coding exons in total corresponding to 150 kb of genomic DNA) of these genes makes routine molecular diagnostics costly and time-consuming. The road to this 'holy grail' of diagnosis is thus often lengthy and full of pitfalls, and relies on a combination of clinical, biochemical and molecular findings.

Historically, muscle biopsies were the routine and primary step undertaken for diagnostic purposes, and double immunostaining with a basement-membrane marker enabled recognition of ColVI deficiency in patients with UCMD 63, but not in patients with BM. The current diagnostic method of determining ColVI involvement is primarily based on immunocytochemistry of cultured skin fibroblasts, but this analysis is only available in a limited number of laboratories to date. A number of antibodies recognizing human ColVI are now commercially available and may be used for such techniques; in particular, the refined protocol proposed by Hicks et al.64, using a polyclonal antibody raised against mature ColVI from human placenta, has better sensitivity, especially in fibroblast cultures from patients with BM (Figure 3). The absence or alteration of ColVI secretion in cultured fibroblasts, associated with clinical symptoms compatible with a diagnosis of ColVI myopathy, certainly warrants further genetic analysis.

<p>Figure 3</p>

Collagen type VI (ColVI) expression study in cultured skin fibroblasts

Collagen type VI (ColVI) expression study in cultured skin fibroblasts. (A) Representative images obtained using the protocol from 67 in which ColVI (red) is labeled with monoclonal antibody MAB1944 (Chemicon (now Millipore), Billerica, MA, USA) and perlecan (green) with monoclonal antibody MAB1948 (Chemicon). Note that ColVI expression appeared clearly altered in a patient with an early severe (ES) form and less so in patients with intermediate (Int) or Bethlem myopathy (BM) forms, compared with control fibroblasts (Cont). (B) Using the protocol of Hicks et al. 64, which detects ColVI (red) with polyclonal antibody Ab6588 (Abcam, Cambridge, UK) and fibronectin (green) with monoclonal antibody F15 (Sigma Chemical Co., St Louis, MO, USA), the sensitivity of the method is increased, and defective ColVI secretion could be detected in all patients' samples. Insets indicate nuclei, labeled using DAPI. Bars are 50 μm. (Images courtesy of Corine Gartioux and Valérie Allamand).

Over the past decade, the development of genetic studies has demonstrated the heterogeneity and complexity of the molecular mechanisms at play in ColVI myopathies. An autosomal recessive pattern of inheritance was initially thought to be involved in UCMD, and linkage analysis led to the identification of mutations in the COL6A2 and COL6A3 genes 656667. However, numerous dominant de novo mutations have now been shown to be involved, accounting for more than 50% of the mutations causing UCMD 38394268. Similarly, autosomal dominant mutations were first identified in the COL6A1 and COL6A2 genes in families with BM, suggesting that BM was mostly familial and inherited as an autosomal dominant disease 69, although rare de novo mutations and autosomal recessive mutations have now been reported 70717273. To date, over 200 mutations have been identified in these genes, mostly distributed in the COL6A1 and COL6A2 genes. The most common types of mutations are point mutations, and mutations leading to premature termination codons (PTCs) and exon skipping (Figure 4). Among the former, missense changes affecting glycine residues in the triple helical domains of the corresponding proteins are the most common, and are often dominant de novo. Because these changes affect crucial amino acids within the collagenous domains, they hamper triple-helix formation 74757677. Splice mutations resulting in in-frame exon skipping are generally dominant de novo mutations, and exons 16 of COL6A3 and 14 of COL6A1 seem to be preferentially affected, leading to UCMD or BM phenotypes, respectively 5375787980818283. Nonsense mutations and small deletions or insertions inducing PTCs within the coding frame are mostly inherited as recessive mutations, and lead to loss of function of the protein 42536875767980818283848586878889909192. These mutations are responsible for most UCMD phenotypes. Nevertheless, it should be noted that genotype-phenotype correlations are very difficult to identify.

<p>Figure 4</p>

Repartition of the various types of mutations identified in the COL6A1, COL6A2 and COL6A3 genes

Repartition of the various types of mutations identified in the COL6A1, COL6A2 and COL6A3 genes. This schematic reflects, to the best of our ability, the distribution of 258 allelic mutations (98 on COL6A1, 113 on COL6A2 and 47 on COL6A3). Dominant de novo mutations represent 67% of the missense mutations, affecting glycine residues in the TH domains (Gly in TH), 57% of small deletions (< 5 amino acids) and 44% of splice-site mutations leading to in-frame exon skipping, whereas 97% of mutations leading to premature termination codons (PTCs) are familial (recessive or dominant).

It has recently been shown that all types of mutations alter transcript levels, and that in the case of PTC-bearing transcripts, which are specifically degraded via the nonsense-mediated mRNA decay (NMD 9394) pathway, quantification of the three COL6A mRNAs is a helpful tool to pinpoint the mutated gene, thereby facilitating these cumbersome molecular analyses 42. The NMD-induced degradation of PTC-bearing transcripts may also, at least in part, explain why the parents of patients with UCMD who themselves harbor recessive mutations are asymptomatic; their heterozygous status sustains the expression of 50% of the 'normal' protein, thereby leading to a 'functional loss of heterozygosity'. The study by Briñas and collaborators also provided some genotype-phenotype correlations in a cohort of patients with early-onset ColVI myopathy, showing that recessive mutations leading to PTC were associated with severe phenotypes 42. Genetic studies are further complicated by a possibly variable penetrance as reported by Peat et al. 89.

Finally, the highly polymorphic nature of the COL6A genes makes it difficult to definitely assign pathogenicity to some variants, especially missense ones that do not affect glycine residues within the triple-helix domains of the proteins. In addition, these 'polymorphisms' may very well play a role in the extreme clinical variability of these conditions, particularly in patients carrying identical mutations but presenting with variable severity.

The types of mutations identified also reflect the methods used in laboratories performing these analyses (for example, sequencing of genomic DNA or of the coding sequences on cDNA), but the emergence of high-throughput methods (arrays) is likely to allow the identification of as yet unknown or under-recognized pathogenic mechanisms, such as large gene rearrangements, or promoter or deep intronic mutations, as recently illustrated in two reports 9596.

Animal models and pathophysiology

Limited access to muscle biopsies hinders extensive investigations of the specific cellular mechanisms leading to the development of the muscle pathology, and in vitro/ex vivo cellular systems only partially reproduce the complexity of the tissue. The development of the first animal model of ColVI deficiency in 1998, engineered by invalidating the Col6a1 gene in mice, has proven central to understanding the cellular pathways involved in these diseases. Homozygous animals were reported to develop a mild myopathic phenotype, and were initially described as a model of BM 97. Interestingly, the diaphragm was the most affected muscle, with signs of necrosis evidenced by uptake of Evan's blue dye 97. Subsequently, a latent mitochondrial dysfunction accompanied by ultrastructural alterations of mitochondria and the sarcoplasmic reticulum, resulting in spontaneous apoptosis, was found in about one-third of muscle fibers 98. Reduced contractile strength of the diaphragm and other muscle groups was also reported in Col6a1-/- mice in this initial study 98. The maximal isometric tension generated by ColVI-deficient skinned fibers from gastrocnemius was found to be reduced in a recent report; however, using a protocol of eccentric contractions in vivo, no muscle force drop was found, indicating that the lack of ColVI does not impair myofibrillar function 99. Importantly, mitochondrial dysfunction was also reported in cultured muscle cells from patients and could be reversed by cyclosporin (Cs)A, an immunosuppressive drug that prevents the opening of the mitochondrial permeability transition pore through binding to cyclophilin D, and also inhibits the phosphatase calcineurin 100101. Another in vitro study showed that patients-derived skin fibroblasts behave differently from myoblasts in that respect, and also questioned the specificity of this mitochondrial dysfunction 102, warranting further studies on the matter.

A role for cell survival had previously been proposed for ColVI because it was shown to prevent anti-α1 integrin-mediated apoptosis and trigger the downregulation of bax, a pro-apoptotic molecule 103104.

Recently, a study of the autophagic process in muscles of Col6a1 knockout mice revealed that autophagy was not induced efficiently 105. The ensuing defective autophagy provides the link between the previously described mitochondrial dysfunction and myofiber degeneration, as abnormal organelles and molecules cannot be efficiently cleared from the cell. This study further showed that the forced induction of autophagy, either by dietary restriction or by treatment with rapamycin or CsA, ameliorated the phenotype of the Col6a1-/- mice (Figure 5). A similar alteration of autophagy was also detected in muscle biopsies derived from nine patients with UCMD or BM 105. These data thus provide a basis for novel therapeutic targets to promote the elimination of defective organelles in ColVI-deficient skeletal muscle.

<p>Figure 5</p>

Current pathological hypotheses and therapeutic targets

Current pathological hypotheses and therapeutic targets. The currently known cascade of main events leading to myofiber degeneration in ColVI-deficient skeletal muscle is shown. Mitochondrial dysfunction (due in part to the defective permeability transition pore (PTP) opening) triggers an energetic imbalance with the increased levels of phosphorylated adenosine monophosphate-activated protein kinase (p-AMPK), Ca2+ overload and the production of reactive oxygen species (ROS). Lack of autophagy induction exacerbates the cellular dysfunction because defective mitochondria and proteins (such as p62 aggregates) are not cleared from the cytoplasm. Together, these defects lead to increased apoptosis. Potential therapeutic interventions are indicated in green.

Morpholino-mediated knock-down of the col6a1 and col6a3 genes in zebrafish embryos showed that collagen VI deficiency significantly impairs muscle development and function 106. Increased apoptosis, partially prevented by CsA treatment, was also described in the zebrafish morphants 106. As in other instances, perturbation of muscle components leads to a more severe phenotype in zebrafish than in mouse models, which may in part be due to intrinsic differences in muscle development in these species, especially in terms of timing. Zebrafish models have emerged as major in vivo models of neuromuscular disorders, and seem to be particularly well suited for whole-organism screens for potential pharmacological treatments, as recently illustrated in zebrafish models of Duchenne muscular dystrophy 107.

Therapeutic intervention

To date, no curative treatment exists for these disorders, and most patients rely on supportive treatment of symptoms, usually involving orthopedic (spinal deformations, contractures) and respiratory complications 108.

The unveiling of mitochondrial dysfunction led to an open pilot trial in five patients with UCMD or BM treated orally with CsA for 1 month 109. This study reported normalization of the mitochondrial dysfunction and decrease of apoptosis of muscle cells following this short-term treatment 110111. Longer treatment (up to 2 years) had some beneficial effect on muscle function in these patients but did not prevent progression of the disease in the children 38. Debio-025 (D-MeAla3EtVal4-cyclosporin; DebioPharm) prevents the inappropriate opening of the mitochondrial permeability transition pore (PTP) without interfering with calcineurin 112, and was shown to restore mitochondrial function in cultured muscle cells of patients 100 and in Col6a1-/- mice 113. Debio-025 is currently being tested in a phase II clinical trial in patients with chronic hepatitis C. Another anti-apoptotic pharmacological agent that is being investigated in the context of ColVI myopathies is Omigapil (N-(dibenz(b, f)oxepin-10-ylmethyl)-N-methyl-N-prop-2-ynylamine maleate; Santhera Pharmaceuticals), a chemical derivative of (-)-deprenyl, which was shown to reduce GAPDH-Siah1-mediated apoptosis in a mouse model of laminin α2 chain deficiency 114.

It should be noted that translating some of this research from animal models to patients represents a challenging task, particularly because, to date, these drugs have not been approved for use in children, the patient population with the most severe forms of ColVI myopathies. In addition, there is concern about these therapeutic approaches because of the pleiotropic, and potentially harmful, consequences of anti-apoptotic and/or pro-autophagy treatments. Furthermore, such approaches aiming at modulating downstream pathways would not address the primary defect in these disorders, that is, lack of ColVI in the connective tissue, and would thus need to be continually administered. For the sake of discussion, several alternative, and not necessarily exclusive, therapeutic avenues that would sustain re-expression of ColVI may be envisioned. These approaches may consist of gene-based therapies, such as vector delivery of ColVI-coding sequence, and antisense inhibition of mutant transcripts exerting dominant-negative effects 96. Additionally, as nonsense mutations leading to PTCs are often associated with early-onset, severe phenotypes 42, pharmacological approaches aiming to 'force' translation of PTCs (a phenomenon known as 'translational readthrough' 115) may prove beneficial for a subset of patients carrying these types of mutations. However, the complex assembly process and regulation of ColVI may prove challenging and may limit the realistic options to be investigated.

Conclusions

The past decade of research on neuromuscular disorders has proven very exciting, and has seen ColVI myopathies emerge as an important set of disorders, rather under-recognized until recently. Many challenges remain despite the tremendous advances in the understanding of their genetic, biochemical and pathophysiological bases. It is hoped that the decade(s) to come will see the development of safe and efficient therapies for these disorders. Consequently, as for other rare diseases, the scientific community, and patient organizations, and patients and their families have become increasingly aware of the need for databases, both clinical and genetic, to facilitate recruitment of patients for upcoming clinical trials.

List of abbreviations used

BM: Bethlem myopathy; ColVI: collagen type VI; COL6A: gene(s) encoding the alpha chain(s) of collagen VI; CsA: cyclosporin A; DAPI: 4',6-diamidino-2-phenylindole; EDMD: Emery-Dreifuss muscular dystrophy; EDS: Ehlers-Danlos syndrome; LMNA: gene encoding lamin A/C; MDC1A: congenital muscular dystrophy with laminin α2 chain deficiency; PTP: permeability transition pore; MRI: magnetic resonance imaging; PTC: premature termination codon; ROS: reactive oxygen species; SEPN1: gene encoding selenoprotein N; TNXB: gene encoding tenascin-X; UCMD: Ullrich congenital muscular dystrophy

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

All authors contributed to the writing of this manuscript. All authors read and approved the final manuscript.

Authors' information

Valérie Allamand holds a PhD in Human Genetics, and currently leads a group focusing on ColVI myopathies in the research unit directed by Thomas Voit. In 2009, she co-organized, with Drs Kate Bushby and Luciano Merlini, the 166th ENMC International Workshop on Collagen Type VI-Related Myopathies (22-24 May 2009, Naarden, The Netherlands). She is the genetic curator of the UMD-COL6 databases, developed in the context of the Treat-NMD European network of excellence.

Laura Briñas obtained her PhD in Molecular Biology. She joined the Institut de Myologie in September 2006 as a post-doctoral fellow, and has been involved in the molecular analysis of mutations in the genes encoding collagen VI and the dissection of their cellular consequences.

Susana Quijano-Roy is a child neurologist with previous medical training in La Paz Hospital (Madrid, Spain) and Boston Children's Hospital (USA). She has held a clinical practice since 2001 at Garches Neuromuscular Reference Center (GNMH), and leads the pediatric EMG laboratory at Necker Enfants Hospital, Paris. She obtained her PhD in 2004 with a study on congenital muscular dystrophies (CMDs). She is part or the international expert group that is currently defining diagnosis, standards of care and natural history of CMDs and establishing outcome measures for future therapeutic trials.

Tanya Stojkovic is a medical doctor, specialized in neurophysiology and neuromuscular disorders. She has worked in the neuromuscular clinical unit directed by Professor Eymard since 2006 (Pitié-Salpêtière, Institut de Myologie, Paris, France). She is involved, as a clinician, in the diagnosis of neuromuscular disorders. She has a special interest in ColVI-related myopathies.

Pascale Richard holds a PharmD, Graduation from Medical Biologist and a PhD in molecular genetics. She is head of the 'Functional Unit of Molecular Cardiogenetics and Myogenetics' at Pitié-Salpêtrière Hospital in Paris, where she developed a functional unit focused on the molecular diagnosis of cardiomyopathies and congenital and progressive myopathies in close collaboration with the research units. This unit constitutes the only laboratory in France where the genetic diagnosis of ColVI myopathies is proposed.

Gisèle Bonne holds a PhD in developmental physiology, followed by a post-doctoral training in human genetics. She currently leads the team 'Genetics and Pathophysiology of Neuromuscular Disorders' in the research unit directed by Thomas Voit at the Institut de Myologie in Paris. Her research group focuses on neuromuscular disorders caused by mutations in the lamin A/C gene.

Acknowledgements

We thank Corine Gartioux and Céline Ledeuil for excellent technical skills. This study was funded by the Institut National de la Santé et de la Recherche Médicale (Inserm), Association Française contre les Myopathies (AFM), UPMC Université Paris 06, Centre National de la Recherche Scientifique (CNRS), Assistance Publique-Hôpitaux de Paris (AP-HP). We apologize to all colleagues who might have not been cited in this review due to space limitations.

<p>Mammalian collagen receptors</p>LeitingerBHohenesterEMatrix Biol20072614615510.1016/j.matbio.2006.10.00717141492<p>Binding of the proteoglycan decorin to collagen type VI</p>BidansetDGuidryCRosenbergLChoiHTimplRHookMJ Biol Chem1992267525052561544908<p>Structural and functional features of the alpha 3 chain indicate a bridging role for chicken collagen VI in connective tissues</p>BonaldoPRussoVBucciottiFDolianaRColombattiABiochemistry1990291245125410.1021/bi00457a0212322559<p>Binding of the NG2 proteoglycan to type VI collagen and other extracellular matrix molecules</p>BurgMATilletETimplRStallcupWBJ Biol Chem1996271261102611610.1074/jbc.271.42.261108824254<p>Corneal cell-matrix interactions: type VI collagen promotes adhesion and spreading of corneal fibroblasts</p>DoaneKJYangGBirkDEExp Cell Res199220049049910.1016/0014-4827(92)90200-R1572410<p>The collagen family members as cell adhesion proteins</p>HeinoJBioEssays2007291001101010.1002/bies.2063617876790<p>Ultrastructure of type VI collagen in human skin and cartilage suggests an anchoring function for this filamentous network</p>KeeneDEngvallEGlanvilleRJ Cell Biol19881071995200610.1083/jcb.107.5.199521153163182942<p>Type VI collagen microfibrils: evidence for a structural association with hyaluronan</p>KieltyCMWhittakerSPGrantMEShuttleworthCAJ Cell Biol199211897999010.1083/jcb.118.4.97922895771323568<p>Type VI collagen anchors endothelial basement membranes by interacting with Type IV collagen</p>KuoH-JMaslenCLKeeneDRGlanvilleRWJ Biol Chem1997272265222652910.1074/jbc.272.42.265229334230<p>Interaction of intact type VI collagen with hyaluronan</p>McDevittCAMarcelinoJTuckerLFEBS Letters199129416717010.1016/0014-5793(91)80660-U1756855<p>Integrin and Arg-Gly-Asp dependence of cell adhesion to the native and unfolded triple helix of collagen type VI</p>PfaffMAumailleyMSpecksUKnolleJZerwesHGTimplRExp Cell Res199320616717610.1006/excr.1993.11348387021<p>Structure of recombinant N-terminal globule of type VI collagen alpha 3 chain and its binding to heparin and hyaluronan</p>SpecksUMayerUNischtRSpissingerTMannKTimplREngelJChuMLEmbo J199211428142905570011425570<p>Keratan sulfate and dermatan sulfate proteoglycans associate with type VI collagen in fetal rabbit cornea</p>TakahashiTChoHIKublinCLCintronCJ Histochem Cytochem1993411447145710.1177/41.10.82454048245404<p>The membrane-spanning proteoglycan NG2 binds to collagens V and VI through the central nonglobular domain of its core protein</p>TilletERuggieroFNishiyamaAStallcupWBJ Biol Chem1997272107691077610.1074/jbc.272.16.107699099729<p>Complexes of matrilin-1 and biglycan or decorin connect collagen VI microfibrils to both collagen II and aggrecan</p>WibergCKlattARWagenerRPaulssonMBatemanJFHeinegardDMorgelinMJ Biol Chem2003278376983770410.1074/jbc.M30463820012840020<p>Sequence analysis of alpha 1(VI) and alpha 2(VI) chains of human type VI collagen reveals internal triplication of globular domains similar to the A domains of von Willebrand factor and two alpha 2(VI) chain variants that differ in the carboxy terminus</p>ChuMLPanTCConwayDKuoHJGlanvilleRWTimplRMannKDeutzmannREMBO J19898193919464010542551668<p>Mosaic structure of globular domains in the human type VI collagen alpha 3 chain: similarity to von Willebrand factor, fibronectin, actin, salivary proteins and aprotinin type protease inhibitors</p>ChuMLZhangRZPanTCStokesDConwayDKuoHJGlanvilleRMayerUMannKDeutzmannRTimplREMBO J199093853935516781689238<p>Cloning and chromosomal localization of human genes encoding the three chains of type VI collagen</p>WeilDMatteiMGPassageEN'GuyenVCPribula-ConwayDMannKDeutzmannRTimplRChuMLAm J Hum Genet19884243544517151623348212<p>Three novel collagen VI Chains, {alpha}4(VI), {alpha}5(VI), and {alpha}6(VI)</p>FitzgeraldJRichCZhouFHHansenUJ Biol Chem2008283201702018010.1074/jbc.M71013920018400749<p>Three novel collagen vi chains with high homology to the {alpha}3 chain</p>GaraSKGrumatiPUrciuoloABonaldoPKobbeBKochMPaulssonMWagenerRJ Biol Chem2008283106581067010.1074/jbc.M70954020018276594<p>The knee osteoarthritis susceptibility locus DVWA on chromosome 3p24.3 is the 5' part of the split COL6A4 gene</p>WagenerRGaraSKKobbeBPaulssonMZauckeFMatrix Biol20092830731010.1016/j.matbio.2009.05.00319486942<p>Expression of the collagen VI [alpha]5 and [alpha]6 chains in normal human skin and in skin of patients with collagen VI-related myopathies</p>SabatelliPGaraSKGrumatiPUrciuoloAGualandiFCurciRSquarzoniSZamparelliAMartoniEMerliniLPaulssonMBonaldoPWagenerRJ Invest Dermatol20101319910720882040<p>Variants in a novel epidermal collagen gene (col29a1) are associated with atopic dermatitis</p>SöderhällCMarenholzIKerscherTRüschendorfFEsparza-GordilloJWormMGruberCMayrGAlbrechtMRohdeKSchulzHWahnUHubnerNLeeYAPLoS Biology20075e24210.1371/journal.pbio.0050242197112717850181<p>A pooling-based genome-wide analysis identifies new potential candidate genes for atopy in the European Community Respiratory Health Survey (ECRHS)</p>Castro-GinerFBustamanteMRamon GonzalezJKogevinasMJarvisDHeinrichJAntoJ-MWjstMEstivillXde CidRBMC Medical Genetics20091012810.1186/1471-2350-10-128279750519961619<p>A common variant on chromosome 11q13 is associated with atopic dermatitis</p>Esparza-GordilloJWeidingerSFolster-HolstRBauerfeindARuschendorfFPatoneGRohdeKMarenholzISchulzFKerscherTHubnerUWahnSSchreiberAFrankeRVoglerSHeathHBaurechtNNovakERodriguezTIlligM-ALee-KirschACiechanowiczMKurekTPiskackovaMMacekY-ALee RuetherANat Genet20094159660110.1038/ng.34719349984<p>Distinct regions control transcriptional activation of the alpha1(VI) collagen promoter in different tissues of transgenic mice</p>BraghettaPFabbroCPiccoloSMarvulliDBonaldoPVolpinDBressanGMJ Cell Biol19961351163117710.1083/jcb.135.4.116321333808922394<p>An enhancer required for transcription of the Col6a1 gene in muscle connective tissue is induced by signals released from muscle cells</p>BraghettaPFerrariAFabbroCBizzottoDVolpinDBonaldoPBressanGMExp Cell Res20083143508351810.1016/j.yexcr.2008.08.00618761340<p>Analysis of transcription of the Col6a1 gene in a specific set of tissues suggests a new variant of enhancer region</p>GirottoDFabbroCBraghettaPVitalePVolpinDBressanGMJ Biol Chem2000275173811739010.1074/jbc.M00007520010747869<p>The supramolecular organization of collagen VI microfibrils</p>BaldockCSherrattMJShuttleworthCAKieltyCMJ Mol Biol200333029730710.1016/S0022-2836(03)00585-012823969<p>Structural basis of type VI collagen dimer formation</p>BallSBellaJKieltyCShuttleworthAJ Biol Chem2003278153261533210.1074/jbc.M20997720012473679<p>Amino acid sequence of the triple-helical domain of human collagen type VI</p>ChuMConwayDPanTBaldwinCMannKDeutzmannRTimplRJ Biol Chem198826318601186063198591<p>Structure and macromolecular organization of type VI collagen</p>EngelJFurthmayrHOdermattEVon Der MarkHAumailleyMFleischmajerRTimplRAnnals of the New York Academy of Sciences1985460253710.1111/j.1749-6632.1985.tb51154.x3938630<p>Molecular assembly, secretion, and matrix deposition of type VI collagen</p>EngvallEHessleHKlierGJ Cell Biol198610270371010.1083/jcb.102.3.70321141163456350<p>Electron-microscopical approach to a structural model of intima collagen</p>FurthmayrHWiedemannHTimplROdermattEEngelJBiochem J198321130331111543606307276<p>Collagens, modifying enzymes and their mutations in humans, flies and worms</p>MyllyharjuJKivirikkoKITrends in Genet200420334310.1016/j.tig.2003.11.004<p>Reduction of Lysyl hydroxylase 3 causes deleterious changes in the deposition and organization of extracellular Matrix</p>RisteliMRuotsalainenHSaloAMSormunenRSipiläLBakerNLLamandeSRVimpari-KauppinenLMyllyläRJ Biol Chem2009284282042821110.1074/jbc.M109.038190278887219696018<p>Secretion and assembly of type IV and VI collagens depend on glycosylation of hydroxylysines</p>SipiläLRuotsalainenHSormunenRBakerNLLamandeSRVapolaMWangCSadoYAszodiAMyllyläRJ Biol Chem2007282333813338810.1074/jbc.M70419820017873278<p>166th ENMC International Workshop on Collagen type VI-related Myopathies, 22-24 May 2009, Naarden, The Netherlands</p>AllamandVMerliniLBushbyKNeuromusc Disord20102034635410.1016/j.nmd.2010.02.01220211562<p>Collagen VI related muscle disorders</p>LampeAKBushbyKMDJ Med Genet20054267368510.1136/jmg.2002.002311173612716141002<p>A clinical and histological study of Ullrich's disease (congenital atonic-sclerotic muscular dystrophy)</p>NonakaIUneYIshiharaTMiyoshinoSNakashimaTSugitaHNeuroped19811219720810.1055/s-2008-1059651<p>Kongenitale, atonisch-sklerotische Muskeldystrophie, ein weiteres Typus der heredodegenerativen Erkrankungen des neuromuskulären Systems</p>UllrichOZ Gesamte Neurol Psychiat193012617120110.1007/BF02864097<p>Early onset collagen VI myopathies: genetic and clinical correlations</p>BriñasLRichardPQuijano-RoySGartiouxCLedeuilCMakriSFerreiroAMaugenreSTopalogluHHalilogluGPénisson-BesnierIJeannetP-YMerliniLNavarroCToutainAChaigneDDesguerreIde Die-SmuldersCDunandMEchenneBEymardBKuntzerTMaincentKMayerMPlessisGRivierFRoelensFStojkovicTTaratutoALubienieckiFAnnals of Neurol20106851152010.1002/ana.22087<p>Natural history of Ullrich congenital muscular dystrophy</p>NadeauAKinaliMMainMJimenez-MallebreraCAloysiusAClementENorthBManzurAYRobbSAMercuriEMuntoniFNeurology200973253110.1212/WNL.0b013e3181aae85119564581<p>Benign myopathy with autosomal dominant inheritance: a report of three pedigrees</p>BethlemJVan WijngaardenGKBrain1976999110010.1093/brain/99.1.91963533<p>Bethlem myopathy</p>De VisserMvan der KooiAJJobsisGJMyologyNew York: McGraw-HillFranzini-Amstrong AGEaC200411351146<p>Bethlem myopathy: a slowly progressive congenital muscular dystrophy with contractures</p>JobsisGJBoersJMBarthPGde VisserMBrain199912264965510.1093/brain/122.4.64910219778<p>Ullrich congenital muscular dystrophy: connective tissue abnormalities in the skin support overlap with Ehlers-Danlos syndromes</p>KirschnerJHausserIZouYSchreiberGChristenHJBrownSCAnton-LamprechtIMuntoniFHanefeldFBonnemannCGAm J Med Genet A2005132296301<p>Bethlem myopathy: A study of two families</p>NaliniAGayathriNNeurol India20105866566610.4103/0028-3886.6868420739820<p>Bethlem myopathy (BETHLEM) and Ullrich scleroatonic muscular dystrophy: 100th ENMC International Workshop, 23-24 November 2001, Naarden, The Netherlands</p>PepeGBertiniEBonaldoPBushbyKGiustiBde VisserMGuicheneyPLattanziGMerliniLMuntoniFNishinoINonakaIBen YaouRSabatelliPSewryCTopalogluHvan der KooiANeuromusc Disord20021298499310.1016/S0960-8966(02)00139-612467756<p>Autosomal recessive myosclerosis myopathy is a collagen VI disorder</p>MerliniLMartoniEGrumatiPSabatelliPSquarzoniSUrciuoloAFerliniAGualandiFBonaldoPNeurology2008711245125310.1212/01.wnl.0000327611.01687.5e18852439<p>Novel mutations in collagen VI genes: expansion of the Bethlem myopathy phenotype</p>ScacheriPCGillandersEMSubramonySHVedanarayananVCroweCAThakoreNBinglerMHoffmanEPNeurology20025859360211865138<p>Prevalence of genetic muscle disease in northern England: in-depth analysis of a muscle clinic population</p>NorwoodFLMHarlingCChinneryPFEagleMBushbyKStraubVBrain20091323175318610.1093/brain/awp23619767415<p>Primary collagen VI deficiency is the second most common congenital muscular dystrophy in Japan</p>OkadaMKawaharaGNoguchiSSugieKMurayamaKNonakaIHayashiYKNishinoINeurology2007691035104210.1212/01.wnl.0000271387.10404.4e17785673<p>Diagnosis and etiology of congenital muscular dystrophy</p>PeatRASmithJMComptonAGBakerNLPaceRABurkinDJKaufmanSJLamandeSRNorthKNNeurology20087131232110.1212/01.wnl.0000284605.27654.5a18160674<p>Genetic diseases of connective tissues: cellular and extracellular effects of ECM mutations</p>BatemanJFBoot-HandfordRPLamandeSRNat Rev Genet20091017318319204719<p>Clinical and molecular overlap between myopathies and inherited connective tissue diseases</p>VoermansNCBönnemannCGHuijingPAHamelBCvan KuppeveltTHde HaanASchalkwijkJvan EngelenBGJenniskensGJNeuromusc Disord20081884385610.1016/j.nmd.2008.05.01718818079<p>Neuromuscular involvement in various types of Ehlers-Danlos syndrome</p>VoermansNCvan AlfenNPillenSLammensMSchalkwijkJZwartsMJvan RooijIAHamelBCvan EngelenBGAnnals of Neurol20096568769710.1002/ana.21643<p>Differentiating Emery-Dreifuss muscular dystrophy and collagen VI-related myopathies using a specific CT scanner pattern</p>DeconinckNDionEBen YaouRFerreiroAEymardBBriñasLPayanCVoitTGuicheneyPRichardPAllamandVBonneGStojkovicTNeuromusc Disord20102051752310.1016/j.nmd.2010.04.00920576434<p>Muscle magnetic resonance imaging involvement in muscular dystrophies with rigidity of the spine</p>MercuriEClementsEOffiahAPichiecchioAVascoGBiancoFBerardinelliAManzurAPaneMMessinaSGualandiFRicciERutherfordMMuntoniFAnnals of Neurol20106720120810.1002/ana.21846<p>Muscle magnetic resonance imaging in patients with congenital muscular dystrophy and Ullrich phenotype</p>MercuriECiniCPichiecchioAAllsopJCounsellSZolkipliZMessinaSKinaliMBrownSCJimenezCNeuromusc Disord20031355455810.1016/S0960-8966(03)00091-912921792<p>Muscle MRI in Ullrich congenital muscular dystrophy and Bethlem myopathy</p>MercuriELampeAAllsopJKnightRPaneMKinaliMBonnemannCFlaniganKLapiniIBushbyKPepeGMuntoniFNeuromusc Disord20051530331010.1016/j.nmd.2005.01.00415792870<p>Muscle ultrasound in Bethlem myopathy</p>BönnemannCBrockmannKHanefeldFNeuroped200334335336<p>Reduced cell anchorage may cause sarcolemma-specific collagen VI deficiency in Ullrich disease</p>KawaharaGOkadaMMoroneNIbarraCANonakaINoguchiSHayashiYKNishinoINeurology2007691043104910.1212/01.wnl.0000271386.89878.2217785674<p>A refined diagnostic algorithm for Bethlem myopathy</p>HicksDLampeAKBarresiRCharltonRFiorilloCBonnemannCGHudsonJSuttonRLochmullerHStraubVBushbyKNeurology2008701192119910.1212/01.wnl.0000307749.66438.6d18378883<p>Ullrich scleroatonic muscular dystrophy is caused by recessive mutations in collagen type VI</p>Camacho VanegasOBertiniEZhangR-ZPetriniSMinosseCSabatelliPGiustiBChuM-FPepeGProc Natl Acad Sci USA2001987516752110.1073/pnas.1210275983470011381124<p>Collagen VI status and clinical severity in Ullrich congenital muscular dystrophy: phenotype analysis of 11 families linked to the COL6 loci</p>DemirEFerreiroASabatelliPAllamandVMakriSEchenneBMaraldiNMMerliniLTopalogluHGuicheneyPNeuroped200435103112<p>Mutations in COL6A3 cause severe and mild phenotypes of Ullrich congenital muscular dystrophy</p>DemirESabatelliPAllamandVFerreiroAMoghadaszadehBMakreloufMTopalogluHEchenneBMerliniLGuicheneyPAm J Hum Genet2002701446145810.1086/34060841999111992252<p>Dominant collagen VI mutations are a common cause of Ullrich congenital muscular dystrophy</p>BakerNLMorgelinMPeatRGoemansNNorthKNBatemanJFLamandeSRHum Mol Genet20051427929315563506<p>Type VI collagen mutations in Bethlem myopathy, an autosomal dominant myopathy with contractures</p>JobsisGJKeizersHVreijlingJPde VisserMSpeerMCWoltermanRABaasFBolhuisPANat Genet19961411311510.1038/ng0996-1138782832<p>Autosomal recessive inheritance of classic Bethlem myopathy</p>FoleyARHuYZouYColumbusAShoffnerJDunnDMWeissRBBonnemannCGNeuromusc Disord20091981381710.1016/j.nmd.2009.09.010278790619884007<p>Autosomal recessive Bethlem myopathy</p>GualandiFUrciuoloAMartoniESabatelliPSquarzoniSBovolentaMMessinaSMercuriEFranchellaAFerliniABonaldoPMerliniLNeurology2009731883189110.1212/WNL.0b013e3181c3fd2a19949035<p>A novel de novo mutation in the triple helix of the COL6A3 gene in a two-generation Italian family affected by Bethlem myopathy. A diagnostic approach in the mutations' screening of type VI collagen</p>PepeGBertiniEGiustiBBrunelliTComeglioPSaittaBMerliniLChuMLFedericiGAbbateRNeuromusc Disord1999926427110.1016/S0960-8966(99)00014-010399756<p>Bethlem myopathy (BETHLEM) 86th ENMC international workshop, 10-11 November 2000, Naarden, The Netherlands</p>PepeGde VisserMBertiniEBushbyKVanegasOCChuMLLattanziGMerliniLMuntoniFUrtizbereaANeuromusc Disord20021229630510.1016/S0960-8966(01)00275-911801404<p>Kinked collagen VI tetramers and reduced microfibril formation as a result of Bethlem myopathy and introduced triple helical glycine mutations</p>LamandéSRMorgelinMSelanCJobsisGJBaasFBatemanJFJ Biol Chem20022771949195610.1074/jbc.M10993220011707460<p>Bethlem myopathy and engineered collagen VI triple helical deletions prevent intracellular multimer assembly and protein secretion</p>LamandéSRShieldsKAKornbergAJShieldLKBatemanJFJ Biol Chem1999274218172182210.1074/jbc.274.31.2181710419498<p>Collagen VI glycine mutations: perturbed assembly and a spectrum of clinical severity</p>PaceRPeatRBakerNZamursLMörgelinMIrvingMAdamsNBatemanJMowatDSmithNLamontPMooreSMathewsKNorthKLamandéSAnnals of Neurol20086429430310.1002/ana.21439<p>A Bethlem myopathy Gly to Glu mutation in the von Willebrand factor A domain N2 of the collagen alpha3(VI) chain interferes with protein folding</p>SasakiTHohenesterEZhangRZGottaSSpeerMCTandanRTimplRChuMLFaseb J20001476176810744632<p>Molecular consequences of dominant Bethlem myopathy collagen VI mutations</p>BakerNLMörgelinMPaceRAPeatRAAdamsNEGardnerRJMRowlandLPMillerGDe JonghePCeulemansBHannibalMCEdwardsMThompsonEMJacobsonRQuinlivanRCMAftimosSKornbergAJNorthKNBatemanJFLamandéSRAnnals of Neurol20079999NA<p>Automated genomic sequence analysis of the three collagen VI genes: applications to Ullrich congenital muscular dystrophy and Bethlem myopathy</p>LampeAKDunnDMvon NiederhausernACHamilCAoyagiALavalSHMarieSKChuM-LSwobodaKMuntoniFBönnemannCGFlaniganKMBushbyKMDWeissRBJ Med Genet20054210812010.1136/jmg.2004.023754173600015689448<p>Exon skipping mutations in collagen VI are common and are predictive for severity and inheritance</p>LampeAKZouYSudanoDO'BrienKKHicksDLavalSHCharltonRJimenez-MallebreraCZhangRZFinkelRSTennekoonGSchreiberGvan der KnaapMSMarksHStraubVFlaniganKMChuMLMuntoniFBushbyKMDBönnemannCGHuman Mut20082980982210.1002/humu.20704<p>Detection of common and private mutations in the COL6A1 gene of patients with Bethlem myopathy</p>LucioliSGiustiBMercuriECamacho VanegasOLucariniLPietroniVUrtizbereaJ-ABen YaouRde VisserMvan der KooiAJBönnemannCGIannacconeSTMerliniLBushbyKMuntoniFBertiniEChuMLPepeGNeurology2005641931193710.1212/01.WNL.0000163990.00057.6615955946<p>New molecular mechanism for Ullrich congenital muscular dystrophy: a heterozygous in-frame deletion in the COL6A1 gene causes a severe phenotype</p>PanTZhangRSudanoDMarieSBönnemannCChuMAm J Hum Genet20037335536910.1086/377107118037212840783<p>A Heterozygous splice site mutation in COL6A1 leading to an in-frame deletion of the [alpha]1(VI) collagen chain in an Italian family affected by Bethlem myopathy</p>PepeGGiustiBBertiniEBrunelliTSaittaBComeglioPBologneseAMerliniLFedericiGAbbateRChuM-LBiochem Biophys Res Com199925880280710.1006/bbrc.1999.068010329467<p>Dominant and recessive COL6A1 mutations in Ullrich scleroatonic muscular dystrophy</p>GiustiBLucariniLPietroniVLucioliSBandinelliBPetriniSGartiouxCTalimBRoelensFMerliniLTopalogluHBertiniEGuicheneyPPepeGAnn Neurol20055840041010.1002/ana.2058616130093<p>A comparative analysis of collagen VI production in muscle, skin and fibroblasts from 14 Ullrich congenital muscular dystrophy patients with dominant and recessive COL6A mutations</p>Jimenez-MallebreraCMaioliMAKimJBrownSCFengLLampeAKBushbyKHicksDFlaniganKMBonnemannCSewryCAMuntoniFNeuromusc Disorders20061657158210.1016/j.nmd.2006.07.01516935502<p>The role of the alpha3(VI) chain in collagen VI assembly. Expression of an alpha3(VI) chain lacking N-terminal modules N10-N7 restores collagen VI assembly, secretion, and matrix deposition in an alpha3(VI)- deficient cell line</p>LamandéSRSigalasEPanTCChuMLDziadekMTimplRBatemanJFJ Biol Chem19982737423743010.1074/jbc.273.13.74239516440<p>A homozygous COL6A2 intron mutation causes in-frame triple-helical deletion and nonsense-mediated mRNA decay in a patient with Ullrich congenital muscular dystrophy</p>LucariniLGiustiBZhangR-ZPanTCJimenez-MallebreraCMercuriEMuntoniFPepeGChuM-LHum Genet200511746046610.1007/s00439-005-1318-816075202<p>Identification and characterization of novel collagen VI non-canonical splicing mutations causing ullrich congenital muscular dystrophy</p>MartoniEUrciuoloASabatelliPFabrisMBovolentaMNeriMGrumatiPD'AmicoAPaneMMercuriEBertiniEMerliniLBonaldoPFerliniAGualandiFHuman Mut200930E662E67210.1002/humu.21022<p>Variable penetrance of COL6A1 null mutations: Implications for prenatal diagnosis and genetic counselling in Ullrich congenital muscular dystrophy families</p>PeatRABakerNLJonesKJNorthKNLamandeSRNeuromusc Disorders20071754755710.1016/j.nmd.2007.03.01717537636<p>COL6A1 genomic deletions in Bethlem myopathy and Ullrich muscular dystrophy</p>PepeGLucariniLZhangRZPanTGiustiBQuijano-RoySGartiouxCBushbyKMGuicheneyPChuMLAnn Neurol20065919019510.1002/ana.2070516278855<p>Collagen VI microfibril formation is abolished by an alpha(VI) von Willebrand factor A-domain mutation in a patient with Ullrich congenital muscular dystrophy</p>TooleyLDZamursLKBeecherNBakerNLPeatRAAdamsNEBatemanJFNorthKNBaldockCLamandeSRJ Biol Chem2010<p>Recessive COL6A2 C-globular missense mutations in Ullrich congenital muscular dystrophy</p>ZhangR-ZZouYPanT-CMarkovaDFertalaAHuYSquarzoniSReedUCMarieSKNBonnemannCGChuM-LJ Biol Chem2010285100051001510.1074/jbc.M109.093666284316420106987<p>The nonsense-mediated decay RNA surveillance pathway</p>ChangYFImamJSWilkinsonMFAnnual Review of Biochemistry200776517410.1146/annurev.biochem.76.050106.09390917352659<p>Quality control of mRNA function</p>MaquatLECarmichaelGGCell200110417317610.1016/S0092-8674(01)00202-111207359<p>Identification of a deep intronic mutation in the COL6A2 gene by a novel custom oligonucleotide CGH array designed to explore allelic and genetic heterogeneity in collagen VI-related myopathies</p>BovolentaMNeriMMartoniEUrciuoloASabatelliPFabrisMGrumatiPMercuriEBertiniEMerliniLBonaldoPFerliniAGualandiFBMC Medical Genetics20101144285089520302629<p>Large genomic deletions: A novel cause of Ullrich congenital muscular dystrophy</p>FoleyARHuYZouYYangMMedneLLeachMConlinLKSpinnerNShaikhTHFalkMNeumeyerAMBlissLTsengBSWinderTLBönnemannCGAnnals of Neurol20116920621110.1002/ana.22283<p>Collagen VI deficiency induces early onset myopathy in the mouse: an animal model for Bethlem myopathy</p>BonaldoPBraghettaPZanettiMPiccoloSVolpinDBressanGMHum Mol Genet199872135214010.1093/hmg/7.13.21359817932<p>Mitochondrial dysfunction and apoptosis in myopathic mice with collagen VI deficiency</p>IrwinWBergaminNSabatelliPReggianiCMegighianAMerliniLBraghettaPColumbaroMVolpinDBressanGBernardiPBonaldoPNat Genet20033536737110.1038/ng127014625552<p>Eccentric contractions lead to myofibrillar dysfunction in muscular dystrophy</p>BlaauwBAgateaLTonioloLCanatoMQuartaMDyarKADanieli-BettoDBettoRSchiaffinoSReggianiCJ Applied Physiology2010108105111<p>Mitochondrial dysfunction in the pathogenesis of Ullrich congenital muscular dystrophy and prospective therapy with cyclosporins</p>AngelinATiepoloTSabatelliPGrumatiPBergaminNGolfieriCMattioliEGualandiFFerliniAMerliniLMaraldiNMBonaldoPBernardiPProc Natl Acad Sci USA200710499199610.1073/pnas.0610270104178342717215366<p>Dysfunction of mitochondria and sarcoplasmic reticulum in the pathogenesis of collagen VI muscular dystrophies</p>BernardiPBonaldoPAnnals of the New York Academy of Sciences2008114730331110.1196/annals.1427.00919076452<p>Cyclosporine. A treatment for Ullrich congenital muscular dystrophy: a cellular study of mitochondrial dysfunction and its rescue</p>HicksDLampeAKLavalSHAllamandVJimenez-MallebreraCWalterMCMuntoniFQuijano-RoySRichardPStraubVLochmullerHBushbyKMDBrain200913214715519015158<p>Type VI collagen increases cell survival and prevents anti-[beta]1integrin-mediated apoptosis</p>HowellSJDoaneKJExp Cell Res199824123024110.1006/excr.1998.40519633532<p>Soluble collagen VI drives serum-starved fibroblasts through S Phase and prevents apoptosis via down-regulation of Bax</p>RuhlMSahinEJohannsenMSomasundaramRManskiDRieckenEOSchuppanDJ Biol Chem1999274343613436810.1074/jbc.274.48.3436110567413<p>Autophagy is defective in collagen VI muscular dystrophies, and its reactivation rescues myofiber degeneration</p>GrumatiPColettoLSabatelliPCesconMAngelinABertaggiaEBlaauwBUrciuoloATiepoloTMerliniLMaraldiNMBernardiPSandriMBonaldoPNat Med2010161313132010.1038/nm.224721037586<p>Zebrafish models of collagen VI-related myopathies</p>TelferWRBustaASBonnemannCGFeldmanELDowlingJJHum Mol Genet2010192433244410.1093/hmg/ddq126287688820338942<p>Drug screening in a zebrafish model of Duchenne muscular dystrophy</p>KawaharaGKarpfJAMyersJAAlexanderMSGuyonJRKunkelLMProc Natl Acad Sci USA20111085331533610.1073/pnas.1102116108306921521402949<p>Consensus statement on standard of care for congenital muscular dystrophies</p>WangCHBonnemannCGRutkowskiASejersenTBelliniJBattistaVFlorenceJMScharaUSchulerPMWahbiKAloysiusABashROBéroudCBertiniEBushbyKCohnRDConnollyAMDeconinckNDesguerreIEagleMEstournet-MathiaudBFerreiroAFujakAGoemansNIannacconeSTJouinotPMainMMelaciniPMueller-FelberWMuntoniFJ Child Neurol2010251559158110.1177/088307381038192421078917<p>Cyclosporin A corrects mitochondrial dysfunction and muscle apoptosis in patients with collagen VI myopathies</p>MerliniLAngelinATiepoloTBraghettaPSabatelliPZamparelliAFerliniAMaraldiNMBonaldoPBernardiPProc Natl Acad Sci USA20081055225522910.1073/pnas.0800962105227817918362356<p>Collagen VI myopathies: From the animal model to the clinical trial</p>MaraldiNMSabatelliPColumbaroMZamparelliAManzoliFABernardiPBonaldoPMerliniLAdvances in Enzyme Regulation20094919721110.1016/j.advenzreg.2008.12.00919162063<p>Therapy of collagen VI-related myopathies (Bethlem and Ullrich)</p>MerliniLBernardiPNeurotherapeutics2008561361810.1016/j.nurt.2008.08.00419019314<p>The Nonimmunosuppressive cyclosporin analogs NIM811 and UNIL025 display nanomolar potencies on permeability transition in brain-derived mitochondria</p>HanssonMJMattiassonGMånssonRKarlssonJKeepMFWaldmeierPRueggUTDumontJ-MBesseghirKElmérEJournal of Bioenergetics and Biomembranes20043640741315377880<p>The cyclophilin inhibitor Debio 025 normalizes mitochondrial function, muscle apoptosis and ultrastructural defects in Col6a1-/- myopathic mice</p>TiepoloTAngelinAPalmaESabatelliPMerliniLNicolosiLFinettiFBraghettaPVuagniauxGDumontJMBaldariCTBonaldoPBernardiPBritish Journal of Pharmacology20091571045105210.1111/j.1476-5381.2009.00316.x273766319519726<p>Omigapil ameliorates the pathology of muscle dystrophy caused by laminin-{alpha}2 deficiency</p>ErbMMeinenSBarzaghiPSumanovskiLTCourdier-FruhIRueggMAMeierTJ Pharmacol Exp Ther200933178779510.1124/jpet.109.16075419759319<p>Aminoglycoside-induced translational read-through in disease: overcoming nonsense mutations by pharmacogenetic therapy</p>ZingmanLVParkSOlsonTMAlekseevAETerzicAClin Pharmacol Ther2007819910310.1038/sj.clpt.610001217186006