The importance of genetic diagnosis for Duchenne muscular dystrophy =================================================================== * Annemieke Aartsma-Rus * Ieke B Ginjaar * Kate Bushby ## Abstract Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy are caused by mutations in the dystrophin-encoding *DMD* gene. Large deletions and duplications are most common, but small mutations have been found as well. Having a correct diagnosis is important for family planning and providing proper care to patients according to published guidelines. With mutation-specific therapies under development for DMD, a correct diagnosis is now also important for assessing whether patients are eligible for treatments. This review discusses different mutations causing DMD, diagnostic techniques available for making a genetic diagnosis for children suspected of DMD and the importance of having a specific genetic diagnosis in the context of emerging genetic therapies for DMD. * Muscle disease * Diagnosis * Genetics ## Introduction Mutations in the dystrophin-encoding *DMD* gene underlie Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD), a severe (DMD) and milder (BMD) form of inherited, progressive muscle wasting.1 ,2 Normally, the dystrophin protein acts as a shock absorber during muscle fibre contraction by linking the actin of the contractile apparatus to the layer of connective tissue that surrounds each muscle fibre.3 ,4 In DMD, mutations abolish dystrophin function either by disrupting the reading frame or by generating a premature stop codon (figure 1A, B). As a result, the connection between the actin cytoskeleton and connective tissue is lost and muscle fibres are easily damaged during contraction, leading to chronic muscle damage, inflammation and eventually replacement of muscle fibres by fat and fibrotic tissue and thus loss of muscle function.5 ![Figure 1](http://jmg.bmj.com/https://jmg.bmj.com/content/jmedgenet/53/3/145/F1.medium.gif) [Figure 1](http://jmg.bmj.com/content/53/3/145/F1) Figure 1 Schematic depiction of dystrophin transcripts in healthy, Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) individuals. (A) In the normal situation, the dystrophin mRNA consists of 79 exons that are translated into the dystrophin protein. (B) In patients with DMD, protein translation is stopped prematurely. This can be due to frame-shifting mutations (in this example, a deletion of exons 47–50, top panel) that lead to the inclusion of aberrant amino acids and generally premature truncation of translation. Alternatively, a point mutation can change an amino acid codon into a stop codon (bottom panel, nonsense mutation). This premature stop codon will be used instead of the natural stop codon at the end of the transcript. (C) In patients with BMD, mutations maintain the open reading frame (in this example, a deletion of exons 46–54). As such, protein translation does not stop prematurely but continues until the natural stop codon at the end of the mRNA. However, the generated dystrophin will be shorter because it will lack the amino acids encoded by exons 46–54. By contrast, individuals with BMD generally have mutations that maintain the open reading frame, allowing the production of dystrophin proteins with an internal deletion or duplication that can connect actin to the connective tissue, and thus are partially functional (figure 1C). Consequently, individuals with BMD often show a later onset and a slower disease progression, although there is phenotypic variation, with some patients being diagnosed in childhood and others being diagnosed in midlife or later.5 ,6 Almost all DMD patients will develop cardiomyopathy,7 and there is a high risk patients with BMD will develop cardiomyopathy regardless of the skeletal muscle phenotype. The incidence of dystrophinopathies is ∼1 in 4000 (1 in 5000 for DMD and 1 in 20 000 for BMD in male live births).5 ,8–10 Since the *DMD* gene is located on the X-chromosome, DMD and BMD present primarily in males and are maternally inherited. Correct DNA diagnostic analysis is crucial for DMD and BMD patients since it is important for optimal care and family planning, but also provides information on eligibility for mutation-specific treatments.11 In this review, we will explain the different types of mutations that have been reported for the *DMD* gene, discuss genotype–phenotype correlations; outline the genetic diagnosis of children suspected of having DMD; and discuss the importance of a specific genetic diagnosis in the context of emerging genetic therapies. A glossary of terminology is provided in table 1. View this table: [Table 1](http://jmg.bmj.com/content/53/3/145/T1) Table 1 Explaining terminology ## Mutations in the *DMD* gene The *DMD* gene is the largest known human gene, containing 79 exons spanning 2.2 Mb.5 The mutation rate is relatively high; in one in three cases, DMD is caused by a de novo mutation. As such, new cases will arise even with good prenatal diagnostic tools and family counselling for known cases. This high mutation rate also underlies the large variation of mutations that has been identified for patients with DMD (figure 2).6 ,12 Although the majority of patients have a deletion (∼68%) or duplication (∼11%) of one or more exons, small mutations are found as well (∼20% of patients). These deletions and duplications can occur anywhere in the gene, but are concentrated between exons 45–55 and exons 2–10 for deletions and duplications, respectively.13 The outcome of deletion and duplication mutations can be twofold. If the number of nucleotides in the exons that are deleted or duplicated is divisible by 3, the reading frame will not be disrupted. This will allow translation of the mRNA into a dystrophin protein that, although slightly shorter or longer in the centre, contains the N-terminal and C-terminal domains crucial for connecting the actin cytoskeleton to the extracellular matrix. These dystrophins retain some degree of function and are found in patients with BMD. By contrast, when the number of nucleotides of the exons deleted or duplicated is not divisible by 3, the reading frame is shifted, leading to the incorporation of aberrant amino acids into the protein during translation. Often, an incorrect reading frame contains many stop codons, leading to premature termination of translation and production of a truncated protein. The resulting dystrophins lack the crucial domain that connects to the extracellular matrix and are therefore not functional and generally not stable. These mutations are associated with DMD. ![Figure 2](http://jmg.bmj.com/https://jmg.bmj.com/content/jmedgenet/53/3/145/F2.medium.gif) [Figure 2](http://jmg.bmj.com/content/53/3/145/F2) Figure 2 Schematic depiction of the effect of different types of Duchenne muscular dystrophy (DMD)-causing mutations on the dystrophin transcript. (A) Deletions of one or more exons can cause a shift of the open reading frame (in this example, a deletion of exon 45). (B) A duplication of one or more exons can cause a shift of the open reading frame (in this example, a duplication of exon 2). (C) There are several types of small mutations than can cause DMD. Nonsense mutations (top panel) introduce a stop codon prematurely (in this example, the nonsense mutation is located in exon 35). Small insertions or deletions (middle panel) can disrupt the open reading frame (in this example, a 1 bp insertion in exon 35). Finally, mutations affecting the splice sites (bottom panel) generally lead to the exclusion of the affected exon from the mRNA (in this example, exon 43). As such, a single-exon deletion that disrupts the open reading frame is generated on the mRNA level. There are several ways by which small mutations can interfere with dystrophin production (figure 2C). Small deletions or insertions can disrupt the reading frame at an exon level, which like larger out-of-frame deletions or duplications involving one or more exons leads to non-functional dystrophins. Point mutations can convert a codon for an amino acid into a stop codon, which will also result in premature termination of protein translation. Finally, point mutations or small deletions and/or insertions can disrupt a splice site. During the splicing process, introns are removed by the splicing machinery, for which recognition of the splice donor (first two nucleotides of an intron (GU)) and splice acceptor (last two nucleotides of an intron (AG)) is crucial. Generally, an exon for which the acceptor or donor splice site is mutated will no longer be recognised by the splicing machinery, leading to the exon being excluded from the mRNA. Thus, splice-site mutations generally cause a single-exon deletion at the mRNA level, which will lead to a partially functional or a non-functional dystrophin depending on whether the omitted exon was in-frame or out-of-frame, respectively. In <1% of patients, other types of mutations are found; for example, deep intronic mutations can lead to an intronic region being recognised as an exon by the splicing machinery (cryptic or pseudo-exons) leading to its inclusion in the mRNA.6 ,12 Cryptic exons can disrupt the reading frame and/or will contain stop codons preventing the production of functional dystrophin. Missense mutations are rare in patients with DMD, and the few that have been reported are located in the cysteine-rich domain of dystrophin and abolish the binding to β-dystroglycan, which in turn connects to the extracellular matrix.12 ,14 Therefore, in effect these missense mutations are similar to other DMD-causing mutations because they also disrupt the connection between actin cytoskeleton and extracellular matrix. Finally, a handful of cases caused by translocations involving the *DMD* gene have also been reported. These mutations separate one portion of the gene from the other, preventing a full transcript being produced. Notably, translocation mutations involving the *DMD* gene will cause a DMD phenotype in females.15 ,16 Normally, carrier females will be born with 50% of dystrophin positive muscle fibres due to random X-inactivation of the chromosomes carrying the mutated or the normal *DMD* gene. With translocation mutations, however, cells where the translocated *DMD* gene is inactivated will not survive due to a lower dosage of the autosome involved in the translocation. Thus, the unaffected *DMD* gene will always be inactivated and no dystrophin can be produced. ## Genotype–phenotype correlations Close to 10% of genetic mutations do not follow the reading frame rule,6 ,12 that is, patients with in-frame mutations can present with DMD and patients with out-of-frame mutations can present with BMD. Some frequent exceptions to the reading frame rule are discussed below. However, for certain mutations both patients with DMD and patients with BMD have been reported. Furthermore, for patients with BMD and DMD, disease severity can vary for the same mutations, sometimes even within the same family and there is also variation in the extent in which the heart is affected.6 ,7 ,17–19 As such, most likely genetic modifiers play a role as well in determining disease severity. Discussing these is beyond the scope of this review, and we refer interested readers to a recent review paper by Vo and McNally.20 Generally, the location or size of deletions or duplications that lead to out-of-frame mutations do not affect the clinical phenotype; if the reading frame is disrupted, the C-terminal part of the protein cannot be produced and the protein will not be functional. There are, however, three notable exceptions. First, frame-shifting or nonsense mutations before exon 8 can present as BMD in certain cases.21 This is due to the presence of an alternative translation initiation sites in exon 6 or exon 8 that are activated by some mutations (eg, a deletion of exon 2 or a stop mutation in exon 1 activate an alternative translation initiation side in exon 6), but not others (eg, an exon 2 duplication).21 If exons 3–7 are deleted apparently, it varies between patients whether or not the exon 8 alternative translation initiation site is used.22 Second, patients with nonsense mutations in in-frame exons can present with BMD.6 ,23 This can be explained by the fact that the mutation disrupts sequences involved in exon recognition and, consequently, the exon is occasionally skipped, thus bypassing the nonsense mutation but maintaining the reading frame. Third, patients with DMD with mutations flanking exon 44 show a slightly milder phenotype (‘mild DMD’), with, on average, a later loss of ambulation and a slower decline in the distance walked in 6 min compared with other patients.24 ,25 The most likely explanation is low-level spontaneous skipping of exon 44. Indeed, this has been reported in cultured cells from patients with deletions flanking exon 44.26 This suggestion is further supported by the fact that the dystrophin levels in these patients are on average higher than for patients with DMD with other frame-shifting mutations.27 For in-frame mutations, the location and size of mutations can influence disease severity to some extent.6 ,28 In-frame mutations that abolish the extracellular matrix-interacting domain (encoded by exons 64–70) or all actin binding domains (encoded by exons 2–10 and exons 32–45) will not result in functional proteins and are therefore associated with DMD. In-frame deletions affecting the first 10 exons delete the first two actin binding domains, but spare the third one encoded by exons 32–45, typically leading to a ‘severe BMD’ phenotype. Deletions in the hotspot region (exons 45–55) are generally associated with a milder disease presentation. Deletions between exons 10 and 40 are milder still and are sometimes only associated with cramps and myalgia and are sometimes found in asymptomatic individuals.29 Finally, X-linked dilated cardiomyopathy patients have mutations in the *DMD* gene, but present only with a cardiac phenotype in the absence of skeletal muscle problems.5 ,30 Here, dystrophin is produced in skeletal muscle, while it is not produced or not functional in heart. While most carriers do not present with symptoms, some ‘symptomatic carriers’ present with cramps, myalgia or even progressive muscle weakness.31 ,32 Notably, female carriers are prone to develop dilated cardiomyopathy as well. ## Establishing a genetic diagnosis Physicians should suspect DMD when young boys present with impaired muscle function, frequent falls and Gower's sign.11 ,33 Delayed speech is also frequently observed in young boys with DMD, probably due to the absence of a brain isoform of dystrophin. Serum analysis will reveal the presence of elevated muscle enzymes due to leakage into the bloodstream (most notably creatine kinase (CK), but also transaminases such as aspartate transaminase and alanine transaminase). Upon these findings (delayed muscle function, speech delay and high CK), patients are generally referred to neuromuscular specialists, who request genetic analysis of the *DMD* gene to confirm whether the patient has DMD (figure 3). ![Figure 3](http://jmg.bmj.com/https://jmg.bmj.com/content/jmedgenet/53/3/145/F3.medium.gif) [Figure 3](http://jmg.bmj.com/content/53/3/145/F3) Figure 3 Overview of suggested stepwise mutation analysis when suspecting Duchenne muscular dystrophy (DMD). When reporting mutations, it is important to use standard nomenclature as described on the website of the Human Genome Variation Society ([http://www.hgvs.org/mutnomen/](http://www.hgvs.org/mutnomen/)). Furthermore, it is important to include patients in national patient registries (upon consent), so they can be contacted when mutation-specific therapies are tested in clinical trials or become available on the market. The majority of mutations will be detected by multiplex ligation-dependent probe amplification (MLPA) or exon sequencing on DNA level. If no mutation is found, a muscle biopsy can be taken to study dystrophin protein. When dystrophin is abnormal or absent in a muscle biopsy, while no mutation can be found with MLPA or exon sequencing, one can consider analysing muscle RNA to identify the potential inclusion of a cryptic exon due to an intronic mutation (these will not be identified with MLPA or exon sequencing). Given that deletions and duplications of one or more exons are found in the majority (70%) of patients, it is most cost-efficient and labour-efficient to check for these mutations first (see table 2 for the costs of DMD diagnosis in selected European countries). The multiplex ligation-dependent probe amplification (MLPA) analysis is the most reliable test to identify exactly which exons are involved in deletions or duplications.34 This approach uses sets of probes that can hybridise to each of the *DMD* exons. Each set of probes consists of two oligonucleotides that hybridise adjacent to each other to a given exon. Probes have a tail to allow PCR amplification and one of the probes contains a ‘stuffer sequence’, the length of which differs for each pair of probes. Upon hybridisation, a ligation step joins the two probes and a PCR will be performed, which can only amplify ligated probes (ie, probes that were able to hybridise). The resulting PCR fragments are then run for fragment analysis, where each exon is represented by a fragment of a defined length (which varies due to the stuffer sequence). This approach will reveal whether or not a given exon is present and allows the copy number of each exon to be calculated by comparing relative peak heights. MLPA can detect both deletions and duplications in patients and carriers. Small mutations within an exon can prevent binding of the probe to that exon, and as such, these mutations can present as single-exon deletions using the MLPA method.35 Therefore, a PCR using intronic primers is generally used to assess whether the exon is deleted or contains a small mutation. Furthermore, while MLPA detects which exons are involved in the deletion or duplication, it is not informative about the location of the intronic breakpoints. View this table: [Table 2](http://jmg.bmj.com/content/53/3/145/T2) Table 2 Costs for genetic diagnosis of Duchenne muscular dystrophy in selected European countries Alternatively, array comparative genome hybridisation (array CGH) uses probes covering dystrophin exons but also introns conjugated to a glass slide.36 Control and patient DNA is fragmented and labelled with separate fluorophores and hybridised to the probes. Like MLPA, this method will detect the relative abundance of each exon. However, because it also uses probes within introns, it allows pinpointing the location of the intronic breakpoint. Multiplex PCR is not recommended for the genetic diagnosis of dystrophinopathy. However, it provides an alternative, cheaper approach to detecting deletions, and as such, is still used in numerous laboratories.37 ,38 This approach generally uses two primer sets to detect the presence or absence of exons 1, 3, 4, 6, 8, 12, 13, 17, 19, 43, 44, 45, 47, 48, 50, 51, 52 and 60. As such, it is able to pick up the majority of deletion mutations in patients with DMD and BMD. However, often the exact boundary of the mutation will be unknown. For example, when exon 45 is absent, this can be either an out-of-frame exon 45 deletion or an in-frame exon 45–46 deletion (because the presence of exon 46 is not assessed with this approach). This has ramifications for detecting whether a mutation is in-frame or out-of-frame, but also for assessing whether a patient is eligible for mutation-specific therapies like exon skipping. As such, the MLPA or array CGH are the recommended approaches to use first.33 In fact, historic cases identified with multiplex PCR in the past for whom the exact boundaries are not known should be reanalysed with MLPA because this can have implications for disease progression (in-frame vs out-of-frame mutations) and for eligibility for exon skipping therapy. If no whole-exon deletions or duplications are found using MLPA, it is still possible that the patient has a small mutation in one of the 79 exons (20% of patients have small mutations). These can be identified using Sanger sequencing of individual exons, which is labour intensive and more expensive than MLPA or array CGH. Protocols for MLPA and Sanger sequencing for the *DMD* gene can be found on the Eurogentest website ([http://www.eurogentest.org](http://www.eurogentest.org)). It should be noted that not all laboratories will follow-up with exon analysis when MLPA (or multiplex PCR analysis) are negative. A recent survey was conducted among 41 individuals (primarily (paediatric) neurologists and clinical geneticists from Europe, Turkey and India) working with patients with DMD and attending a TREAT-NMD DMD masterclass (funded by an educational grant from PTC therapeutics) (survey results are being prepared for publication on [http://www.treat-nmd.eu](http://www.treat-nmd.eu)). This revealed that all respondents were aware of the need for a genetic diagnosis. However, when deletion and duplication tests are negative, >10% of responders would not perform confirmative tests, while close to 90% would, using either exon sequencing or dystrophin analysis on a biopsy followed by exon sequencing. The main reason for not pursuing genetic analysis when deletion and duplication tests are negative was the costs involved in the Sanger sequencing or the lack of funding for shipping samples to laboratories able to conduct the small mutation analysis. It is possible that in the future MLPA, array CGH and Sanger sequencing will be replaced by next-generation sequencing techniques such as whole-exome sequencing for the genetic diagnosis of DMD. In fact, targeted sequencing of the *DMD* gene has already been reported using a library with probes covering the complete *DMD* gene to enrich for DMD and allowing a single-step diagnosis for deletions, duplications and small mutations in patients with DMD and BMD and carriers.39–41 However, currently it is more cost efficient to perform MLPA or array CGH followed by Sanger sequencing when no mutations are found. Obviously, there will be patients for whom no mutations are found. This can be because the mutation is located within an intron, as is the case for <0.5% of reported mutations, although these mutations may be under-represented because they are hard to detect. These mutations will be picked up with next-generation sequencing approaches, although without RNA analysis it may be challenging to assess whether intronic mutations lead to alterations on RNA level. Alternatively, the absence of a dystrophin mutation may indicate that the patient does not have DMD or BMD, but has a different form of muscular dystrophy. Historically, the first step in diagnosing DMD was often protein analysis on a muscle biopsy by immunohistochemistry and/or western blot analysis.42–44 These techniques can reveal the size (western blot), location (immunohistochemistry) and abundance (both) of dystrophin, but in a diagnostic setting are most frequently used to show the presence or absence of dystrophin. Some laboratories perform a biopsy when MLPA analysis does not reveal deletions or duplications to confirm a dystrophinopathy before embarking on laborious small mutation analysis. However, because a genetic diagnosis is still required when absence of dystrophin is shown in a muscle biopsy, and because a muscle biopsy is an invasive procedure, the standards of care for DMD diagnosis suggest bypassing a muscle biopsy and using only genetic testing to diagnose DMD (figure 3). For most patients, muscle biopsies are never required. In exceptional cases, protein analysis can reveal whether dystrophin is absent in patients with a clear DMD phenotype but with no mutation in the *DMD* gene. If so, RNA can be isolated from the biopsied tissue and analysis may reveal an aberrant dystrophin mRNA transcript (eg, the inclusion of a cryptic exon due to a deep intronic mutation). Muscle biopsies may also be useful in patients who present with a discordant phenotype; for example, if a patient has an in-frame mutation but presents with DMD, a biopsy can reveal the absence of dystrophin. Vice versa, low levels of dystrophin detected in the muscle biopsy of a patient with an out-of-frame mutation presenting with a milder phenotype can explain this discrepancy. However, while dystrophin analysis may provide an explanation for why a disease course is slower or faster than expected, knowing this will not actually change the disease course. As such, this analysis is optional and not mandatory. The standards of care for DMD were published in 2010 and timely genetic diagnosis are part of this.33 ,45 Early diagnosis for DMD is important because having genetic confirmation of the disease-causing mutation has important implications for the family.11 Once a mutation is identified, carrier analysis can be performed for the mother. Carriers are mainly asymptomatic though there is a risk of cardiac involvement, which requires monitoring and intervention if abnormalities are detected. Confirming carrier status in the mother (ie, heterozygosity for a disease-causing mutation) affects family planning and means that her daughters, sisters, cousins and aunts are potentially at risk of being a carrier as well. It is important to realise that even when the mother is not a carrier, it is still possible she has germline mosaicism for the mutation, and as such, is still at risk of having a second son with DMD.46 With the emergence of genetic therapies for DMD, a specific genetic diagnosis has become especially important to evaluate and select each patient's therapeutic options. For example, Translarna (ataluren) has been approved in Europe for the treatment of DMD caused by nonsense mutations (in ambulatory patients aged 5 years or older).47 Given that DMD is a progressive disease and muscle loss is most likely irreversible, such disease-modifying treatments should be initiated as early as possible in eligible patients. Identification of the exact genetic mutation is also important for antisense-mediated exon skipping therapies, which are being tested in clinical trials and for which marketing authorisation applications are ongoing.12 ,48 ## Concluding remarks DMD and BMD are both caused by mutations in the *DMD* gene. Having genetic confirmation of the mutation is important for patients because it has implications for disease prognosis, genetic counselling and evaluating each patient's eligibility for emerging genetic therapies. Given that the progressive muscle wasting is irreversible, it is important that patients are identified as early as possible to consider all potentially effective treatments early in the disease course. On average, patients are being diagnosed at age 4.1 years in an expert centre compared with 4.5 years in 2000.49 Raising awareness with family physicians and other healthcare professionals who see young children could improve this further. Finally, given that new genetic therapies are emerging, one could consider offering neonatal screening for DMD. ## Footnotes * Contributors AA-R and KB discussed the general outline of the manuscript. AA-R wrote the first draft of the manuscript and drafted the figures and tables and finalised the final version. IBG and KB provided comments and suggestions to the manuscript and discussed the outline of the figures. * Competing interests AA-R declares being employed by LUMC, which has patents on exon skipping. As co-inventor of some of these patents, AA-R is entitled to a share of royalties. AAR is on the advisory board of ProQR and has been an ad hoc consultant for GLC consultancy, Global Guidepoint and PTC Therapeutics. Remuneration for these activities is paid to LUMC. KB declares having been or being on the advisory board of Acceleron, AVI Biopharma and Santhera Pharmaceuticals, being on the steering committee of BioMarin Nederland and being or having been a consultant for Debiopharm, Genzyme, GSK, Prosensa Therapeutics/BioMarin Pharmaceutical, PTC Therapeutics, Lilly Pharmaceuticals, Pfizer, Summit Corporation, Insight Research Group, Galapogos SASU, Shire Human Genetic Therapies Inc, Amsterdam Molecular Therapeutics, European Neuromuscular Centre, Bristol-Myers Squibb Company and Solid Ventures LLC. * Provenance and peer review Not commissioned; externally peer reviewed. * Data sharing statement The paper refers to a questionnaire done among neurologists and clinical geneticists. This work is currently prepared for online preparation. The results are shared here with the reviewers and the editor. This is an Open Access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: [http://creativecommons.org/licenses/by-nc/4.0/](http://creativecommons.org/licenses/by-nc/4.0/) ## References 1. Emery AE. The muscular dystrophies. Lancet 2002;359:687–95. [doi:10.1016/S0140-6736(02)07815-7](http://dx.doi.org/10.1016/S0140-6736(02)07815-7) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1016/S0140-6736(02)07815-7&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=11879882&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) [Web of Science](http://jmg.bmj.com/lookup/external-ref?access_num=000174107400029&link_type=ISI) 2. Flanigan KM. Duchenne and Becker muscular dystrophies. Neurol Clin 2014;32:671–88, viii. [doi:10.1016/j.ncl.2014.05.002](http://dx.doi.org/10.1016/j.ncl.2014.05.002) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1016/j.ncl.2014.05.002&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=25037084&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 3. Koenig M, Monaco AP, Kunkel LM. The complete sequence of dystrophin predicts a rod-shaped cytoskeletal protein. Cell 1988;53:219–28. [doi:10.1016/0092-8674(88)90383-2](http://dx.doi.org/10.1016/0092-8674(88)90383-2) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1016/0092-8674(88)90383-2&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=3282674&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) [Web of Science](http://jmg.bmj.com/lookup/external-ref?access_num=A1988N126600007&link_type=ISI) 4. Aartsma-Rus A. Dystrophin analysis in clinical trials. J Neuromuscul Dis2014;1:41–53. 5. Muntoni F, Torelli S, Ferlini A. Dystrophin and mutations: one gene, several proteins, multiple phenotypes. Lancet Neurol 2003;2:731–40. [doi:10.1016/S1474-4422(03)00585-4](http://dx.doi.org/10.1016/S1474-4422(03)00585-4) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1016/S1474-4422(03)00585-4&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=14636778&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) [Web of Science](http://jmg.bmj.com/lookup/external-ref?access_num=000186665800016&link_type=ISI) 6. Aartsma-Rus A, Van Deutekom JC, Fokkema IF, van Ommen GJ, den Dunnen JT. Entries in the Leiden Duchenne muscular dystrophy mutation database: an overview of mutation types and paradoxical cases that confirm the reading-frame rule. Muscle Nerve 2006;34:135–44. [doi:10.1002/mus.20586](http://dx.doi.org/10.1002/mus.20586) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1002/mus.20586&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=16770791&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) [Web of Science](http://jmg.bmj.com/lookup/external-ref?access_num=000239397800002&link_type=ISI) 7. Birnkrant DJ, Ararat E, Mhanna MJ. Cardiac phenotype determines survival in Duchenne muscular dystrophy. Pediatr Pulmonol 2015. http://dx.doi.org/10.1002/ppul.23215 8. Mendell JR, Shilling C, Leslie ND, Flanigan KM, al-Dahhak R, Gastier-Foster J, Kneile K, Dunn DM, Duval B, Aoyagi A, Hamil C, Mahmoud M, Roush K, Bird L, Rankin C, Lilly H, Street N, Chandrasekar R, Weiss RB. Evidence-based path to newborn screening for Duchenne muscular dystrophy. Ann Neurol 2012;71:304–13. [doi:10.1002/ana.23528](http://dx.doi.org/10.1002/ana.23528) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1002/ana.23528&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=22451200&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 9. Moat SJ, Bradley DM, Salmon R, Clarke A, Hartley L. Newborn bloodspot screening for Duchenne muscular dystrophy: 21 years experience in Wales (UK). Eur J Hum Genet 2013;21:1049–53. [doi:10.1038/ejhg.2012.301](http://dx.doi.org/10.1038/ejhg.2012.301) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1038/ejhg.2012.301&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=23340516&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 10. Helderman-van den Enden AT, Madan K, Breuning MH, van der Hout AH, Bakker E, de Die-Smulders CE, Ginjaar HB. An urgent need for a change in policy revealed by a study on prenatal testing for Duchenne muscular dystrophy. Eur J Hum Genet 2013;21:21–6. [doi:10.1038/ejhg.2012.101](http://dx.doi.org/10.1038/ejhg.2012.101) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1038/ejhg.2012.101&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=22669413&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 11. Abbs S, Tuffery-Giraud S, Bakker E, Ferlini A, Sejersen T, Mueller CR. Best practice guidelines on molecular diagnostics in Duchenne/Becker muscular dystrophies. Neuromuscul Disord 2010;20:422–7. [doi:10.1016/j.nmd.2010.04.005](http://dx.doi.org/10.1016/j.nmd.2010.04.005) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1016/j.nmd.2010.04.005&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=20466545&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 12. Bladen CL, Salgado D, Monges S, Foncuberta ME, Kekou K, Kosma K, Dawkins H, Lamont L, Roy AJ, Chamova T, Guergueltcheva V, Chan S, Korngut L, Campbell C, Dai Y, Wang J, Barisic N, Brabec P, Lahdetie J, Walter MC, Schreiber-Katz O, Karcagi V, Garami M, Viswanathan V, Bayat F, Buccella F, Kimura E, Koeks Z, van den Bergen JC, Rodrigues M, Roxburgh R, Lusakowska A, Kostera-Pruszczyk A, Zimowski J, Santos R, Neagu E, Artemieva S, Rasic VM, Vojinovic D, Posada M, Bloetzer C, Jeannet PY, Joncourt F, Diaz-Manera J, Gallardo E, Karaduman AA, Topaloglu H, El SR, Stringer A, Shatillo AV, Martin AS, Peay HL, Bellgard MI, Kirschner J, Flanigan KM, Straub V, Bushby K, Verschuuren J, Aartsma-Rus A, Beroud C, Lochmuller H. The TREAT-NMD DMD global database: analysis of more than 7,000 Duchenne muscular dystrophy mutations. Hum Mutat 2015;36:395–402. [doi:10.1002/humu.22758](http://dx.doi.org/10.1002/humu.22758) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1002/humu.22758&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=25604253&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 13. Ankala A, Kohn JN, Hegde A, Meka A, Ephrem CL, Askree SH, Bhide S, Hegde MR. Aberrant firing of replication origins potentially explains intragenic nonrecurrent rearrangements within genes, including the human DMD gene. Genome Res 2012;22:25–34. [doi:10.1101/gr.123463.111](http://dx.doi.org/10.1101/gr.123463.111) [Abstract/FREE Full Text](http://jmg.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ2Vub21lIjtzOjU6InJlc2lkIjtzOjc6IjIyLzEvMjUiO3M6NDoiYXRvbSI7czoyNDoiL2ptZWRnZW5ldC81My8zLzE0NS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 14. Vulin A, Wein N, Strandjord DM, Johnson EK, Findlay AR, Maiti B, Howard MT, Kaminoh YJ, Taylor LE, Simmons TR, Ray WC, Montanaro F, Ervasti JM, Flanigan KM. The ZZ domain of dystrophin in DMD: making sense of missense mutations. Hum Mutat 2014;35:257–64. [doi:10.1002/humu.22479](http://dx.doi.org/10.1002/humu.22479) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1002/humu.22479&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=24302611&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 15. Ray PN, Belfall B, Duff C, Logan C, Kean V, Thompson MW, Sylvester JE, Gorski JL, Schmickel RD, Worton RG. Cloning of the breakpoint of an X;21 translocation associated with Duchenne muscular dystrophy. Nature 1985;318:672–5. [doi:10.1038/318672a0](http://dx.doi.org/10.1038/318672a0) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1038/318672a0&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=3001530&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 16. Trippe H, Wieczorek S, Kotting J, Kress W, Schara U. Xp21/A translocation: a rarely considered genetic cause for manifesting carriers of duchenne muscular dystrophy. Neuropediatrics 2014;45:333–5. [doi:10.1055/s-0034-1383824](http://dx.doi.org/10.1055/s-0034-1383824) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1055/s-0034-1383824&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=25046452&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 17. Ferreiro V, Giliberto F, Muniz GM, Francipane L, Marzese DM, Mampel A, Roque M, Frechtel GD, Szijan I. Asymptomatic Becker muscular dystrophy in a family with a multiexon deletion. Muscle Nerve 2009;39:239–43. [doi:10.1002/mus.21193](http://dx.doi.org/10.1002/mus.21193) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1002/mus.21193&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=19012301&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) [Web of Science](http://jmg.bmj.com/lookup/external-ref?access_num=000262837700017&link_type=ISI) 18. van den Bergen JC, Wokke BH, Janson AA, van Duinen SG, Hulsker MA, Ginjaar HB, Van Deutekom JC, Aartsma-Rus A, Kan HE, Verschuuren JJ. Dystrophin levels and clinical severity in Becker muscular dystrophy patients. J Neurol Neurosurg Psychiatry 2014;85:747–53. [doi:10.1136/jnnp-2013-306350](http://dx.doi.org/10.1136/jnnp-2013-306350) [Abstract/FREE Full Text](http://jmg.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiam5ucCI7czo1OiJyZXNpZCI7czo4OiI4NS83Lzc0NyI7czo0OiJhdG9tIjtzOjI0OiIvam1lZGdlbmV0LzUzLzMvMTQ1LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 19. Barp A, Bello L, Politano L, Melacini P, Calore C, Polo A, Vianello S, Soraru G, Semplicini C, Pantic B, Taglia A, Picillo E, Magri F, Gorni K, Messina S, Vita GL, Vita G, Comi GP, Ermani M, Calvo V, Angelini C, Hoffman EP, Pegoraro E. Genetic modifiers of Duchenne muscular dystrophy and dilated cardiomyopathy. PLoS ONE 2015;10:e0141240. [doi:10.1371/journal.pone.0141240](http://dx.doi.org/10.1371/journal.pone.0141240) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1371/journal.pone.0141240&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=26513582&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 20. Vo AH, McNally EM. Modifier genes and their effect on Duchenne muscular dystrophy. Curr Opin Neurol 2015;28:528–34. [doi:10.1097/WCO.0000000000000240](http://dx.doi.org/10.1097/WCO.0000000000000240) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1097/WCO.0000000000000240&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=26263473&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 21. Wein N, Vulin A, Falzarano MS, Szigyarto CA, Maiti B, Findlay A, Heller KN, Uhlen M, Bakthavachalu B, Messina S, Vita G, Passarelli C, Gualandi F, Wilton SD, Rodino-Klapac LR, Yang L, Dunn DM, Schoenberg DR, Weiss RB, Howard MT, Ferlini A, Flanigan KM. Translation from a DMD exon 5 IRES results in a functional dystrophin isoform that attenuates dystrophinopathy in humans and mice. Nat Med 2014;20:992–1000. [doi:10.1038/nm.3628](http://dx.doi.org/10.1038/nm.3628) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1038/nm.3628&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=25108525&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 22. Winnard AV, Mendell JR, Prior TW, Florence J, Burghes AH. Frameshift deletions of exons 3–7 and revertant fibers in Duchenne muscular dystrophy: mechanisms of dystrophin production. Am J Hum Genet 1995;56:158–66. [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=7825572&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) [Web of Science](http://jmg.bmj.com/lookup/external-ref?access_num=A1995QC10100020&link_type=ISI) 23. Ginjaar IB, Kneppers AL, Meulen JD, Anderson LV, Bremmer-Bout M, Van Deutekom JC, Weegenaar J, den Dunnen JT, Bakker E. Dystrophin nonsense mutation induces different levels of exon 29 skipping and leads to variable phenotypes within one BMD family. Eur J Hum Genet 2000;8:793–6. [doi:10.1038/sj.ejhg.5200535](http://dx.doi.org/10.1038/sj.ejhg.5200535) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1038/sj.ejhg.5200535&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=11039581&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) [Web of Science](http://jmg.bmj.com/lookup/external-ref?access_num=000089938600011&link_type=ISI) 24. Bergen JC, Ginjaar I, Niks E, Aartsma-Rus A, Verschuuren J. Prolonged ambulation in Duchenne patients with a mutation amenable to exon 44 skipping. J Neuromuscul Dis 2015;1:91–4. 25. Pane M, Mazzone ES, Sormani MP, Messina S, Vita GL, Fanelli L, Berardinelli A, Torrente Y, D'Amico A, Lanzillotta V, Viggiano E, D'Ambrosio P, Cavallaro F, Frosini S, Bello L, Bonfiglio S, Scalise R, De SR, Rolle E, Bianco F, Van der Haawue M, Magri F, Palermo C, Rossi F, Donati MA, Alfonsi C, Sacchini M, Arnoldi MT, Baranello G, Mongini T, Pini A, Battini R, Pegoraro E, Previtali SC, Napolitano S, Bruno C, Politano L, Comi GP, Bertini E, Morandi L, Gualandi F, Ferlini A, Goemans N, Mercuri E. 6 Minute walk test in Duchenne MD patients with different mutations: 12 month changes. PLoS ONE 2014;9:e83400. [doi:10.1371/journal.pone.0083400](http://dx.doi.org/10.1371/journal.pone.0083400) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1371/journal.pone.0083400&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=24421885&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 26. Aartsma-Rus A, Janson AA, Kaman WE, Bremmer-Bout M, den Dunnen JT, Baas F, van Ommen GJ, Van Deutekom JC. Therapeutic antisense-induced exon skipping in cultured muscle cells from six different DMD patients. Hum Mol Genet 2003;12:907–14. [doi:10.1093/hmg/ddg100](http://dx.doi.org/10.1093/hmg/ddg100) [Abstract/FREE Full Text](http://jmg.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiaG1nIjtzOjU6InJlc2lkIjtzOjg6IjEyLzgvOTA3IjtzOjQ6ImF0b20iO3M6MjQ6Ii9qbWVkZ2VuZXQvNTMvMy8xNDUuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 27. Anthony K, Arechavala-Gomeza V, Ricotti V, Torelli S, Feng L, Janghra N, Tasca G, Guglieri M, Barresi R, Armaroli A, Ferlini A, Bushby K, Straub V, Ricci E, Sewry C, Morgan J, Muntoni F. Biochemical characterization of patients with in-frame or out-of-frame DMD deletions pertinent to exon 44 or 45 skipping. JAMA Neurol 2014;71:32–40. [doi:10.1001/jamaneurol.2013.4908](http://dx.doi.org/10.1001/jamaneurol.2013.4908) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1001/jamaneurol.2013.4908&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=24217213&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 28. Beggs AH, Hoffman EP, Snyder JR, Arahata K, Specht L, Shapiro F, Angelini C, Sugita H, Kunkel LM. Exploring the molecular basis for variability among patients with Becker muscular dystrophy: dystrophin gene and protein studies. Am J Hum Genet 1991;49:54–67. [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=2063877&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) [Web of Science](http://jmg.bmj.com/lookup/external-ref?access_num=A1991FW51100008&link_type=ISI) 29. Gospe SM Jr., Lazaro RP, Lava NS, Grootscholten PM, Scott MO, Fischbeck KH. Familial X-linked myalgia and cramps: a nonprogressive myopathy associated with a deletion in the dystrophin gene. Neurology 1989;39:1277–80. [doi:10.1212/WNL.39.10.1277](http://dx.doi.org/10.1212/WNL.39.10.1277) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1212/WNL.39.10.1277&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=2677830&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 30. Ferlini A, Sewry C, Melis MA, Mateddu A, Muntoni F. X-linked dilated cardiomyopathy and the dystrophin gene. Neuromuscul Disord 1999;9:339–46. [doi:10.1016/S0960-8966(99)00015-2](http://dx.doi.org/10.1016/S0960-8966(99)00015-2) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1016/S0960-8966(99)00015-2&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=10407857&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) [Web of Science](http://jmg.bmj.com/lookup/external-ref?access_num=000081202300010&link_type=ISI) 31. Lee SH, Lee JH, Lee KA, Choi YC. Clinical and genetic characterization of female dystrophinopathy. J Clin Neurol 2015;11:248–51. [doi:10.3988/jcn.2015.11.3.248](http://dx.doi.org/10.3988/jcn.2015.11.3.248) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.3988/jcn.2015.11.3.248&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=26022459&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 32. Soltanzadeh P, Friez MJ, Dunn D, von NA, Gurvich OL, Swoboda KJ, Sampson JB, Pestronk A, Connolly AM, Florence JM, Finkel RS, Bonnemann CG, Medne L, Mendell JR, Mathews KD, Wong BL, Sussman MD, Zonana J, Kovak K, Gospe SM, Jr., Gappmaier E, Taylor LE, Howard MT, Weiss RB, Flanigan KM. Clinical and genetic characterization of manifesting carriers of DMD mutations. Neuromuscul Disord 2010;20:499–504. [doi:10.1016/j.nmd.2010.05.010](http://dx.doi.org/10.1016/j.nmd.2010.05.010) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1016/j.nmd.2010.05.010&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=20630757&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 33. Bushby K, Finkel R, Birnkrant DJ, Case LE, Clemens PR, Cripe L, Kaul A, Kinnett K, McDonald C, Pandya S, Poysky J, Shapiro F, Tomezsko J, Constantin C. Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and pharmacological and psychosocial management. Lancet Neurol 2010;9:77–93. [doi:10.1016/S1474-4422(09)70271-6](http://dx.doi.org/10.1016/S1474-4422(09)70271-6) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1016/S1474-4422(09)70271-6&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=19945913&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) [Web of Science](http://jmg.bmj.com/lookup/external-ref?access_num=000273199700026&link_type=ISI) 34. Lalic T, Vossen RH, Coffa J, Schouten JP, Guc-Scekic M, Radivojevic D, Djurisic M, Breuning MH, White SJ, den Dunnen JT. Deletion and duplication screening in the DMD gene using MLPA. Eur J Hum Genet 2005;13:1231–4. [doi:10.1038/sj.ejhg.5201465](http://dx.doi.org/10.1038/sj.ejhg.5201465) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1038/sj.ejhg.5201465&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=16030524&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 35. Okizuka Y, Takeshima Y, Awano H, Zhang Z, Yagi M, Matsuo M. Small mutations detected by multiplex ligation-dependent probe amplification of the dystrophin gene. Genet Test Mol Biomarkers 2009;13:427–31. [doi:10.1089/gtmb.2009.0002](http://dx.doi.org/10.1089/gtmb.2009.0002) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1089/gtmb.2009.0002&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=19473085&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 36. Bovolenta M, Neri M, Fini S, Fabris M, Trabanelli C, Venturoli A, Martoni E, Bassi E, Spitali P, Brioschi S, Falzarano MS, Rimessi P, Ciccone R, Ashton E, McCauley J, Yau S, Abbs S, Muntoni F, Merlini L, Gualandi F, Ferlini A. A novel custom high density-comparative genomic hybridization array detects common rearrangements as well as deep intronic mutations in dystrophinopathies. BMC Genomics 2008;9:572. [doi:10.1186/1471-2164-9-572](http://dx.doi.org/10.1186/1471-2164-9-572) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1186/1471-2164-9-572&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=19040728&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 37. Beggs AH, Koenig M, Boyce FM, Kunkel LM. Detection of 98% of DMD/BMD gene deletions by polymerase chain reaction. Hum Genet 1990;86:45–8. [doi:10.1007/BF00205170](http://dx.doi.org/10.1007/BF00205170) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=2253937&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) [Web of Science](http://jmg.bmj.com/lookup/external-ref?access_num=A1990EK79100008&link_type=ISI) 38. Chamberlain JS, Chamberlain JR, Fenwick RG, Ward PA, Caskey CT, Dimnik LS, Bech-Hansen NT, Hoar DI, Richards S, Covone AE, Govanni R, Abbs S, Bentley DR, Bobrow M, Rysiecki G, Ray PN, Boileau C, Junien C, Boehm C, Venne VL, Fujmura FK, Spiga I, Ferrari M, Tedeschi S, Bakker E, Kneppers AL, van Ommen GJ, Jain K, Spector E, Crandall B, Kiuru A, Savontaus ML, Caskey CT, Chamberlain JS, Chamberlain JR, Rysiecki G. Diagnosis of Duchenne and Becker muscular dystrophies by polymerase chain reaction. A multicenter study. JAMA 1992;267:2609–15. [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1001/jama.1992.03480190051030&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=1573747&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) [Web of Science](http://jmg.bmj.com/lookup/external-ref?access_num=A1992HT82300026&link_type=ISI) 39. Wang Y, Yang Y, Liu J, Chen XC, Liu X, Wang CZ, He XY. Whole dystrophin gene analysis by next-generation sequencing: a comprehensive genetic diagnosis of Duchenne and Becker muscular dystrophy. Mol Genet Genomics 2014;289:1013–21. [doi:10.1007/s00438-014-0847-z](http://dx.doi.org/10.1007/s00438-014-0847-z) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1007/s00438-014-0847-z&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=24770780&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 40. Wei X, Dai Y, Yu P, Qu N, Lan Z, Hong X, Sun Y, Yang G, Xie S, Shi Q, Zhou H, Zhu Q, Chu Y, Yao F, Wang J, He J, Yang Y, Liang Y, Yang Y, Qi M, Yang L, Wang W, Wu H, Duan J, Shen C, Wang J, Cui L, Yi X. Targeted next-generation sequencing as a comprehensive test for patients with and female carriers of DMD/BMD: a multi-population diagnostic study. Eur J Hum Genet 2014;22:110–18. [doi:10.1038/ejhg.2013.82](http://dx.doi.org/10.1038/ejhg.2013.82) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1038/ejhg.2013.82&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=23756440&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 41. Lim BC, Lee S, Shin JY, Kim JI, Hwang H, Kim KJ, Hwang YS, Seo JS, Chae JH. Genetic diagnosis of Duchenne and Becker muscular dystrophy using next-generation sequencing technology: comprehensive mutational search in a single platform. J Med Genet 2011;48:731–6. [doi:10.1136/jmedgenet-2011-100133](http://dx.doi.org/10.1136/jmedgenet-2011-100133) [Abstract/FREE Full Text](http://jmg.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToiam1lZGdlbmV0IjtzOjU6InJlc2lkIjtzOjk6IjQ4LzExLzczMSI7czo0OiJhdG9tIjtzOjI0OiIvam1lZGdlbmV0LzUzLzMvMTQ1LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 42. Anderson LV, Davison K. Multiplex Western blotting system for the analysis of muscular dystrophy proteins. Am J Pathol 1999;154:1017–22. [doi:10.1016/S0002-9440(10)65354-0](http://dx.doi.org/10.1016/S0002-9440(10)65354-0) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1016/S0002-9440(10)65354-0&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=10233840&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) [Web of Science](http://jmg.bmj.com/lookup/external-ref?access_num=000079616600010&link_type=ISI) 43. Anthony K, Arechavala-Gomeza V, Taylor LE, Vulin A, Kaminoh Y, Torelli S, Feng L, Janghra N, Bonne G, Beuvin M, Barresi R, Henderson M, Laval S, Lourbakos A, Campion G, Straub V, Voit T, Sewry CA, Morgan JE, Flanigan KM, Muntoni F. Dystrophin quantification: Biological and translational research implications. Neurology 2014;83:2062–9. [doi:10.1212/WNL.0000000000001025](http://dx.doi.org/10.1212/WNL.0000000000001025) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1212/WNL.0000000000001025&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=25355828&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 44. Beekman C, Sipkens JA, Testerink J, Giannakopoulos S, Kreuger D, Van Deutekom JC, Campion GV, de Kimpe SJ, Lourbakos A. A sensitive, reproducible and objective immunofluorescence analysis method of dystrophin in individual fibers in samples from patients with duchenne muscular dystrophy. PLoS ONE 2014;9:e107494. [doi:10.1371/journal.pone.0107494](http://dx.doi.org/10.1371/journal.pone.0107494) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1371/journal.pone.0107494&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=25244123&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 45. Bushby K, Finkel R, Birnkrant DJ, Case LE, Clemens PR, Cripe L, Kaul A, Kinnett K, McDonald C, Pandya S, Poysky J, Shapiro F, Tomezsko J, Constantin C. Diagnosis and management of Duchenne muscular dystrophy, part 2: implementation of multidisciplinary care. Lancet Neurol 2010;9:177–89. [doi:10.1016/S1474-4422(09)70272-8](http://dx.doi.org/10.1016/S1474-4422(09)70272-8) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1016/S1474-4422(09)70272-8&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=19945914&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) [Web of Science](http://jmg.bmj.com/lookup/external-ref?access_num=000273922000015&link_type=ISI) 46. Bakker E, Van BC, Bonten EJ, van de Vooren MJ, Veenema H, Van HW, van Ommen GJ, Vandenberghe A, Pearson PL. Germline mosaicism and Duchenne muscular dystrophy mutations. Nature 1987;329:554–6. [doi:10.1038/329554a0](http://dx.doi.org/10.1038/329554a0) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1038/329554a0&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=2889144&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 47. Haas M, Vlcek V, Balabanov P, Salmonson T, Bakchine S, Markey G, Weise M, Schlosser-Weber G, Brohmann H, Yerro CP, Mendizabal MR, Stoyanova-Beninska V, Hillege HL. European Medicines Agency review of ataluren for the treatment of ambulant patients aged 5 years and older with Duchenne muscular dystrophy resulting from a nonsense mutation in the dystrophin gene. Neuromuscul Disord 2015;25:5–13. [doi:10.1016/j.nmd.2014.11.011](http://dx.doi.org/10.1016/j.nmd.2014.11.011) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1016/j.nmd.2014.11.011&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=25497400&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) 48. Aartsma-Rus A, Fokkema I, Verschuuren J, Ginjaar I, van DJ, van Ommen GJ, den Dunnen JT. Theoretic applicability of antisense-mediated exon skipping for Duchenne muscular dystrophy mutations. Hum Mutat 2009;30:293–9. [doi:10.1002/humu.20918](http://dx.doi.org/10.1002/humu.20918) [CrossRef](http://jmg.bmj.com/lookup/external-ref?access_num=10.1002/humu.20918&link_type=DOI) [PubMed](http://jmg.bmj.com/lookup/external-ref?access_num=19156838&link_type=MED&atom=%2Fjmedgenet%2F53%2F3%2F145.atom) [Web of Science](http://jmg.bmj.com/lookup/external-ref?access_num=000264011700003&link_type=ISI) 49. van Ruiten HJ, Straub V, Bushby K, Guglieri M. Improving recognition of Duchenne muscular dystrophy: a retrospective case note review. Arch Dis Child 2014;99:1074–7. [doi:10.1136/archdischild-2014-306366](http://dx.doi.org/10.1136/archdischild-2014-306366) [Abstract/FREE Full Text](http://jmg.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTI6ImFyY2hkaXNjaGlsZCI7czo1OiJyZXNpZCI7czoxMDoiOTkvMTIvMTA3NCI7czo0OiJhdG9tIjtzOjI0OiIvam1lZGdlbmV0LzUzLzMvMTQ1LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==)