Flyer

Journal of Neurology and Neuroscience

  • ISSN: 2171-6625
  • Journal h-index: 18
  • Journal CiteScore: 4.35
  • Journal Impact Factor: 3.75
  • Average acceptance to publication time (5-7 days)
  • Average article processing time (30-45 days) Less than 5 volumes 30 days
    8 - 9 volumes 40 days
    10 and more volumes 45 days
Awards Nomination 20+ Million Readerbase
Indexed In
  • Open J Gate
  • Genamics JournalSeek
  • The Global Impact Factor (GIF)
  • China National Knowledge Infrastructure (CNKI)
  • Directory of Research Journal Indexing (DRJI)
  • OCLC- WorldCat
  • Proquest Summons
  • Scientific Journal Impact Factor (SJIF)
  • Euro Pub
  • Google Scholar
  • Secret Search Engine Labs
Share This Page

Review Article - (2018) Volume 9, Issue 3

Application of Multiplex PCR for Detection of Duchunne Muscular Dystrophy: A Childhood Neuromuscular Disorder

Gaurava Srivastava and Preeti Srivastava*

Genetic Centre, Government Medical College and Hospital, Chandigarh, India

*Corresponding Author:

Dr. Preeti Srivastava
DNA Analyst,
Genetic Centre, Government Medical College and Hospital,
Chandigarh, India
Tel: 9478216248
E-mail: preetibtbhu@gmail.com

Received date: May 15, 2018; Accepted date: June 19, 2018; Published date: June 22, 2018

Citation: Srivastava G, Srivastava P (2018) Application of Multiplex PCR for Detection of Duchunne Muscular Dystrophy: A Childhood Neurpmuscular Disorder. J Neurol Neurosci Vol.9 No.1:262. doi:10.21767/2171-6625.1000262

Visit for more related articles at Journal of Neurology and Neuroscience

Abstract

Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder that affects 1 in 3,600 - 6,000 males and is caused by mutation in the dystrophin gene. This is neuromuscular disorder with progressive muscle weakness, which predominantly affect males. Till date no absolute cure of the disease is available in clinical practice. Early diagnosis and timely management are requisite for DMD and it enhances the quality of life of patients. Various diagnostic approaches are available but due to accuracy, early detection and non-invasive method molecular tools are most remarkable in recent era. Multiplex PCR has emerged as one of the most convenient tools for screening of DMD in terms of its sensitivity, specificity, accuracy, cost effectiveness and time consumption. The current study emphasizes advantages and shortcomings of multiplex PCR with reference to most of the past studies along with its challenges for DMD detection in detail. Mutation detection is evidently crucial for diagnosis, as well as it may also be significant for future therapeutic purposes. Further research is important to elucidate specific mutation pattern in association with management and therapies of proband.

Keywords

Duchenne muscular dystrophy; Multiplex PCR; Dystrophin gene; Mutation; Therapeutics

Abbreviations: DMD: Duchenne Muscular Dystrophy; m- PCR: multiplex PCR; PCR: Polymerase Chain Reaction; BMD: Becker Muscular Dystrophy; DGC: Dystrophin- Glycoprotein Complex; aCGH: Comparative Genomic Hybridization Microarray; NGS: Next Generation Sequencing; MLPA: Multiplex Ligation-Dependent Probe Amplification; CK: Creatine Kinase; MPS: Massive Parallel Sequencing

Introduction

Duchenne muscular dystrophy is progressive and serious form of childhood muscle wasting with recessive X-linked inheritance pattern, which alone accounts for approximately 80% of all the myopathies [1]. DMD is the most common fatal genetic condition in children [2] with the prevalence of about 1 in 3,600–9,337 live male births worldwide [3,4]. It is neuromuscular disease generally caused by protein truncating mutations in large DMD gene “dystrophin” at Xp21.2 [5,6]. Dystrophin is a vital protein for myofiber function and musclefiber plasma membrane integrity, mutation in this gene diminishes its expression and biological activity [7,8]. Dystrophin gene is the one of the largest of the 30,000 genes and its mutation leads to DMD [9]. It encodes proteins in human genome: 79 exons cover 2.6 million bp and protein product is of size 427 KDa [8].

Literature Review

DMD is principally caused by out of-frame deletions or duplications, which lead to complete loss of protein, whereas BMD is mostly caused by in-frame deletions or duplications, which leads to partly functional protein with altered-size [10]. The difference between the severe DMD and the allelic, milder Becker Muscular Dystrophy (BMD), occurs due to mutations in the DMD gene, which alter the structure or function of dystrophin or prevent any functional dystrophin from being produced. The clinical diagnosis of D/BMD can be confirmed either by the identification of a mutation in the dystrophin gene or by histological analysis of a patient’s affected muscle tissue; however, DNA testing has been found to be more acceptable than muscle biopsy owing to its less invasive nature, minimal side effects and reduced costs [11].

Clinically, the disease is characterized by progressive muscle wasting, leading to loss of ambulation by 8–15 y of age and early death due to complications of respiratory, orthopedic, cardiac, and smooth muscle dysfunction problems [4,12,13]. The instance of mutation in DMD gene is expected to be either due to inheritance of dystrophin lesions from their maternal side in around two thirds of cases or due to spontaneous mutations in around one third of cases [5,14,15]. The dystrophin protein encoded by DMD gene serves as a stabilizing force within muscle structure [16,17]. The muscular dystrophies are varying in age of onset, degree of severity, mode of inheritance and the muscle group that are primarily affected [18,19]. The initial physical symptoms include developmental delays (locomotor, speech and cognitive in some) followed by abnormal gait, enlarged calves, toe-walking and lean backwards to keep their balance, frequent falls and clumsiness, muscle weakness and difficulty running, jumping, and climbing stairs along with positive Gower’s sign [20-22]. In addition to muscle pathology, intellectual impairment of varying degree is present in about 30% of all DMD patients. DMD affected boys are usually wheelchair bound by the age of 13 years due to eventual weakness [4]. Gradually all the muscles of the body including hearts and lungs muscles become very weak and non-functional which leads to the early death in their 3rd decade of life [4]. Along with clinical symptoms, serum creatine kinase (CK) level is also elevated. Since last two decades, analyses of both the DMD gene and dystrophin protein have improved the diagnosis of DMD [5,23,24] (Figure 1).

jneuro-diagnostic

Figure 1: Step-wise diagnostic strategies for determination of DMD.

Knowing the exact mutation in a proband, today one can determine the possibility of a life span upto their 4th decade [5]. In DMD, 65% accounts for deletion, 5-8% for duplication and the remaining for point mutation and small insertions [25]. The most common cause of death is cardiac and respiratory failure but improvements in health care, use of steroids and assisted ventilation have extended their life span [26].

Approximately, 60% of dystrophin mutations are large insertions or deletions that lead to the frame shift errors, whereas approximately 40% are point mutations or small frameshift rearrangements [27,28]. Around one-third of the DMD patients originate through new mutations while the rest are inherited through carrier mothers or arise from germinal mosaicism. For confirmation of DMD 3 major parameters will be observed that are as follows:

Patients have muscle weakness and large calves. The muscle enzyme in the patient’s blood called CK is very high. This means the muscle cells are breaking down. DNA blood test (genetic test) showed a change in patient’s dystrophin gene, if the genetic test is normal, then muscle biopsy is preferred to perform to visualize dystrophin pattern in the muscle cells.

Dystrophin gene

This gene was identified in 1987, and protein produced by it is also called as dystrophin. This gene is one of the most complex and largest genes identified till date with 2.4 kbp size and 79 exons [29]. The location of the gene is at Xp21.2 of human X-chromosome. This is the largest gene covering 2.5 mega bases [30]. This gene contains up to eight alternate promoters. Three promoters located at the 5’ region of dystrophin gene and gives full length transcription product of size 14 kb mRNA [31]. The gene encodes a large protein with 3685 amino acid residues [32] containing an N-terminal actinbinding domain and multiple spectrin repeats (Figure 2A).

jneuro-dystrophin

Figure 2a: Schematic representation of dystrophin protein and its domains (Adapted from Aartsma-Rus et al. [45]).

The encoded protein forms a component of the dystrophinglycoprotein complex (DGC), which bridges the inner cytoskeleton and the extracellular matrix. Dystrophin protein is principally expressed in skeletal, cardiac and smooth muscles and at lesser extent in brain [33] (Figure 2B).

jneuro-glycoprotein

Figure 2b: Dystrophin and the dystrophin-associated glycoprotein complex (DAGC). (Adapted from Douglas and Wood, [105]).

Mutation analysis of DMD

Mutation studies of the dystrophin gene are focused on detecting deletions or duplications of one or more exons, and multiplex PCR that amplifies selected deletion-prone exons has been used as the most efficient method of mutation detection [34,35]. Mutations in dystrophin protein disrupt the open reading frame and prevent the full translation of its protein product, in ~65% of cases [30,36].

Mutations within the dystrophin gene are mainly distributed within two hotspot regions which map towards the central region (encompassing exon 43-51) and 5’ proximal region (encompassing exon 2-19), small deletions and point mutations appear to be evenly distributed across the gene [37]. Depending on mutation type, the expression as well as function of any protein vary, and dystrophin deficiency leads to DMD/BMD [38-41].

Large deletions spanning one or more exons are identified as the cause of the disease in about 65%–70% of DMD/BMD cases; the remaining has point mutations, mainly nonsense and frame-shift mutations (30%) or duplications (6%) [42,43] (Figure 3).

jneuro-mutations

Figure 3: Types of mutations associated with DMD (Adapted from Roberts et al., [106]).

Mutations are either inherited from asymptomatic female carriers (∼70%) or de novo (∼30%) [44]. Thus, in these gross rearrangements, the reading frame rule explains the clinical difference between DMD and BMD at the molecular level [45].

Generally, in case of DMD mutations reading frame is disrupted, whereas in case of BMD it is maintained. Therefore, most of the DMD mutations create premature stop codons, which presumably results in the expression of truncated proteins that lack the dystrophin C-terminus [46].

Use of multiplex PCR for detection of DMD

PCR is one of the most auspicious method; it has been used for detection of various genetic disorders such as DMD, β- thalassemia, cystic fibrosis, PKU, MCADD, sickle cell anemia, hemophilia etc. [34,47-50], particularly multiplex PCR assay [51]. In all the clinical laboratories, PCR and other sequencebased detection methods are being used progressively more and considered as one of the most important research tools.

For DNA based diagnosis of DMD and BMD, multiplex PCR (m-PCR) has established as one of most important tools. Multiplex PCR is a common enrichment technique for targeted massive parallel sequencing (MPS) protocols.

MPS is widely used in biomedical research and clinical diagnostics as the fast and accurate tool for the detection of short genetic variations [52]. More than two sets and up to ten sets of PCR primers are selected for Multiplexing to amplify those regions of the gene that are susceptible to deletion in DMD/BMD.

In these disorders, 70% of cases are due to a dystrophin gene rearrangement, usually partial deletion, which can be detected by Southern blot analysis using cDNA probes [53]. Southern blotting can reveal large dystrophin gene deletions and duplications in males; however, Southern blotting is timeconsuming, requires hazardous reagents, and is limited to only relatively large deletions/duplications [54].

M-PCR amplification is very useful technique for diagnosis of various disorders including DMD. Major advantages of m-PCR include, step-by-step protocol development, critical parameters such as cost and time deduction for experimentation, more accuracy and efficiency, marked detection process specially in case of mutation analysis.

Therefore, its application was studied and reported by many scientists [55,56]. M-PCR also have some drawbacks which hinders during optimization such as poor sensitivity or specificity and/or preferential amplification of certain specific targets depending on different cases. The m-PCR utilizes more than one primer sets and at times it gives spurious amplification products other than the desired target due to primer dimers formation [57,58]. Therefore, the scientist’s primarily focussing to minimize nonspecific interactions during optimization of m-PCR [59].

Results and Discussion

In DMD gene, m-PCR is utilized for amplification of most commonly deleted and hotspot region exons. Various centers are being using PCR multiplex sets of Chamberlain et al. [34] and Beggs et al. [35,60] and Kunkel et al. [61] and also their modifications.

However, these assays do not cover complete gene with all 79 exons as well as promoters, thus end point of all deletions cannot be characterized. With the implementation of m-PCR, 95-98% of deletion mutation can be detected in male patients only [62].

Exon skipping can be used as an important tool for detection of DMD gene and dystrophin mutations. To provide the advantage a large proportion of DMD patient’s exon 51 was chosen as the target for first clinical trial and its removal would benefit the patients [63].

Synthesis of partially functional dystrophins through antisense oligonucleotide (AON)-mediated exon skipping is a therapeutic approach and exon 51 was firstly targeted through the United States regulators [Food and Drug Administration (FDA)] approval [64]. Deletion of one or more exons are common in DMD causing mutations, cases arising from exonic duplications, nonsense mutations, splice motif detects, microinsertions or deletions may be more amenable to exon skipping [63].

M-PCR has been widely used in D/BMD for deletion detection, however it is time-consuming and difficult to cover all of the exons. Besides, approximately 10% patients with duplication mutations may be misdiagnosed [65]. In 2002, multiplex ligation-dependent probe amplification (MLPA) was invented by Schouten [66], which possesses the capacity to quantify all 79 exons in only 2 reaction sets and facilitates the diagnosis of D/BMD.

With MLPA, identification of deletion, duplication as well as point mutation of whole 79 exons are easy and effective. Murugan et al. [67] utilized m-PCR as well as MLPA for mutation detection in 150 cases. With m-PCR, deletions were identified in 103 patients whereas MLPA identified deletions, duplications and nine additional mutations in patients.

Thus, this study reported MLPA as cost-effective and precise tool for diagnosis of DMD in a developing country like India. Verma et al. [68] reported MLPA as most appropriate tool for intragenic deletions as well as carrier status detection over m- PCR. With the advancement of technologies, the recent therapeutics like antisense oligonucleotide therapy are mutation specific and require the knowledge of mutation to select proper oligo for the patient.

For the detection of genomic rearrangements of the DMD gene custom designed oligonucleotide array-CGH platform is a reliable. Gaudio et al. [69] reported the efficacy of array-CGH platform in detecting submicroscopic copy-number changes involving the DMD gene, as well as providing more precise breakpoint identification at high-resolution and with improved sensitivity. Thus, microarray-based genomic analysis has revolutionized cytogenetics [54,70,71].

With development of various tools and techniques, still molecular tool m-PCR is most convenient and appropriate method for the detection of approximately 98 per cent of deletions, which accounts for 65 per cent of all mutations [34,35]. Earlier studies which have been used m-PCR for D/BMD detection are as discussed in Table 1 [72-106].

S. No. Disorder No. of patients Region of study No. of exons studied No. of patient with deletion Longest deletion Additional tests done Age of patients (Years) Max frequency of exon deleted Deletion
Frequency
Reference
1. DMD/BMD 15 Saudi Arabia 26 12 9 exons (45-53) SCPK 2-19 49 & 50 (13.34%)    [72]
2. DMD/BMD 70 Eastern India (Kolkata) 31 46 12 exons (42-53) SCPK 1-16 48 & 49 65.7%  [73]
  DMD/BMD 108 Kolkata 42 67 38 exons (11-48) SCPK, Carrier Detection 3-15 50 62.05%  [74]
3. DMD/BMD 72 Moroccan 18 37 11 exons (43-52) Western Blotting & immunofluorescence 4-32 46 & 47 65%  [75]
4. DMD 69 Northeast India 17 49 9 exons (44-52) SCPK 2-9 50 (14.38%) 74%  [76]
5. DMD/BMD 15 Western Saudi 9 6 - SCPK 5-19 51 (20%) 40%  [77]
6. DMD/BMD 22 Delhi 27 12 9 exons (44-52) Southern hybridization   45 54.54%  [78]
7. DMD/BMD 88 Gujrat 26 65 8 exons (45-52) SCPK, LDH, Myoglobin - 50 73.86%  [79]
8. BMD 347 Mumbai 32 222 11 exons (45-55) SCPK 12-13 45 (76.1%) 89.1%  [80]
9. DMD 10+10 New Delhi 10 1 4 exons SCPK - 49 5%  [81]
10. DMD 180 New Delhi 22 90 8 exons (45-52) MLPA - 45 50%  [82]
11. DMD 25 Western India (Mumbai) 15 18 - serum creatine kinase (CK) and electromyography - 44 & 51 72%  [83]
12. DMD 101 South India 27 44 - SCPK - 50 73%  [84]
13. DMD 66 South India 19 41 - - - 50  62.1%  [85]
14. DMD 160 Northern India 27 103 7 exons
(45-51)
- - - 64.4%  [86]
15. DMD 100 Western India 19 - - - - - 74%  [42]
16. DMD 74 Iran 19 38 -   - - 51.3%  [87]
17. DMD/BMD 29 Korea 20 21 12 exons (44-55) SCPK, MLPA, Carrier detection 0.4-13 45 72.4%  [88]
18. DMD 84 Taiwan 19 11 16 exons
(45-60)
PCR, DHPLC - - 13%  [89]
19. DMD 442 Japan 19 270 - Complementary DNA (cDNA)
and chromosome analysis
- - 61%  [25]
20. DMD/BMD 29 Iran 29 29 - EMG, MLPA 7.49-21.58 - 100%  [90]
21. DMD/BMD - - 26 - - - - - -  [91]
22. DMD/BMD 110 North West Iran 24 63 8 exons (44-51) SCPK - 50 57.3%  [92]
23. DMD/BMD 123 Serbia, Belgrade 18 71 9 exons (45-53) MLPA - 50 57.7%  [93]
24. DMD/BMD 63 Hong Kong 18 44 9 exons (51-60) MLPA - 50 69.8%  [94]
25. DMD 128 Johannesburg, South Africa 24 40   MLPA - - 31%  [95]
26. DMD/BMD 150 Chennai 30 103 10 exons (45-54) MLPA 3-36 49 and 50 68.7%  [67]
27. DMD 211 Pakistan 18 86 - - - 50 40.75  [96]
28. DMD/BMD 167 China - - - - - - 61.7  [97]
29. DMD/BMD 1053 China - - 20 exons (3-22) MLPA - - 86.4%  [98]
30. DMD 105,
86,
34
Singaporean, Japan, and Vietnam - - - - - 50,
49 & 50,
51
40.0%
51.2%
40.0%
 [99]
31. DMD/BMD 202 Thailand 31 99 14 exons MLPA - - 49%  [100]
32. DMD 20 Malaysia 7 14 7 exons (43-46) (49-51) SCPK 0.5-10 49, 50 and 51 60%  [101]
33. DMD 20 Western India (Mumbai) 32 0 - MLPA 2-9 - -  [102]
34. DMD/BMD 121 North India 28 88 22 exons (1-22) Southern hybridization 1.5-5 45 73%  [103]
35. DMD 50 South Western Maharashtra 21 47 11 exons (42-53) - 2-19 52 94%  [104-106]

Table 1: Deletion pattern in DMD/BMD patients of different regions including India.

Conclusion

DMD and BMD are both caused by mutations in the dystrophin (DMD) gene. Genetic screening and 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. With advancement of molecular tools, the diagnosis of DMD/BMD has become more convenient and accurate. Multiplex PCR is one such most important molecular tool which is most commonly used sensitive method has significantly increased detection of small dystrophin gene mutations and made it possible to diagnose approximately 90% of patients with Duchenne muscular dystrophy by DNA analysis. These findings, combined with cost savings and safety issues, provide compelling reasons to consider DNA analysis as the initial diagnostic test for the suspected dystrophindeficient patient. Thus, with use of m-PCR efficient as well as early detection of DMD/BMD is possible which also provides patient’s timely management and care to enhance their quality of life.

22988

References

  1. Beaudet, AL, Boggs AB, Chinault AC, Eng CM, Gaudio D, et al. (2008) Molecular diagnosis of Duchenne/Becker muscular dystrophy: Enhanced detection of dystrophin gene rearrangements by oligonucleotide array-comparative genomic hybridization. Human Mutation 29: 1100-1107.
  2. Hinton VJ, Nereo NE, DeVivo DC, Goldstein E, Stern Y (2001) Selective deficits in verbal working memory associated with a known genetic etiology: The neuropsychological profile of Duchenne muscular dystrophy J Int Neuropsychological Soc 7: 45–54.
  3. Mah JK,Korngut L,Dykeman J,Day L, Pringsheim T, et al. (2014) A systematic review and meta-analysis on the epidemiology of Duchenne and Becker muscular dystrophy Neuromuscul Disord 24(6): 482–491.
  4. Birnkrant DJ, Bushby K, Bann CM, Apkon SD, Blackwell A, et al. (2018) DMD Care Considerations Working Group (2010) Diagnosis and management of Duchenne muscular dystrophy, part 1 Diagnosis and pharmacological and psychosocial management Lancet Neurol 9: 77–93.
  5. Koenig M, Hoffman EP, Bertelson CJ, Monaco AP, Feener C, et al. (1987) Complete cloning of the Duchenne muscular dystrophy (DMD) cDNA and preliminary genomic organization of the DMD gene in normal and affected individuals Cell 50: 509-517.
  6. Blake DJ, Weir A, Newey SE, Davies KE (2002) Function and genetics of dystrophin and dystrophin-related proteins in muscle. Physiol Rev 82(2): 291-329.
  7. Ervasti JM, Campbell KP (1993) A role for the dystrophin-glycoprotein complex as a transmembrane linker between laminin and actin. J Cell Biol 122: 809–823.
  8. Hoffman EP, Brown RH, Jr, Kunkel LM (1987) Dystrophin: The protein product of the Duchenne muscular dystrophy locus. Cell 51: 919–928.
  9. Monaco AP, Neve RL, Colletti-Feener C, Bertelson CJ, Kurnit DM, et al. (1986) Isolation of candidate cDNAs for portions of the Duchenne muscular dystrophy gene. Nature 323(6089): 646–650.
  10. Monaco AP, Bertelson CJ, Liechti-Gallati S, Moser H, Kunkel LM (1988) An explanation for the phenotypic differences between patients bearing partial deletions of the DMD locus. Genomics 2: 90-95.
  11. Taylor PJ, Maroulis S, Mullan GL, Pedersen RL, Baumli A, et al. (2007) Measurement of the clinical utility of a combined mutation detection protocol in carriers of Duchenne and Becker muscular dystrophy. J Med Genet 44: 368-372.
  12. Wong BL, Christopher C (2002) Corticosteroids in Duchenne muscular dystrophy: A reappraisal. J Child Neurol 17: 183–190.
  13. Cohn RD, Campbell KP (2000) Molecular basis of muscular dystrophies. Muscle Nerve 23: 1456-1471.
  14. Bradley DM, Parsons EP (1998) Neonatal screening for Duchenne muscular dystrophy. Semin Neonatol 3: 27–34.
  15. Webb CL (2005) Parents' perspectives on coping with Duchenne muscular dystrophy. Child Care Health Dev 31: 385-396.
  16. Gorospe JR, Hoffman EP (1992) Duchenne muscular dystrophy. Curr Opin Rheumatol 4(6): 794-800.
  17. Matsumura K, Ervasti JM, Ohlendieck K, Kahl SD, Campbell KP (1992) Association of dystrophin-related protein with dystrophin-associated proteins in mdx mouse muscle. Nature 360, 588-591.
  18. Firth M, Gardner-Medwin D, Hosking G, Wilkinson E (1983) Interviews with parents of boys suffering from Duchenne muscular dystrophy. Dev Med Child Neurol 25: s466-71.
  19. Bushby KM (1999) The limb-girdle muscular dystrophies—Multiple genes, multiple mechanisms. Hum Mol Genet 8: 1875–1882.
  20. Parsons EP, Clarke AJ, Bradley DM (2004) Developmental progress in Duchenne muscular dystrophy: Lessons for earlier detection. European Journal of Pediatrics Neurology 8: 145-153.
  21. Ciafaloni E, Fox DJ, Pandya S, Westfield CP, Puzhankara S, et al. (2009) Delayed diagnosis in duchenne muscular dystrophy: data from the Muscular Dystrophy Surveillance, Tracking, and Research Network (MD STARnet). Journal of Pediatrics 155: 380–385.
  22. McGee RB (2013) Utilization and perceptions of healthcare from a national survey of families with Duchene muscular dystrophy. Degree of Master of Science, Graduate School of Public Health, University of Pittsburgh, USA.
  23. Kunkel LM, Hejtmancik JF, Caskey CT, Speer A, Monaco AP, et al. (1986) Analysis of deletions in DNA from patients with Becker and Duchenne muscular dystrophy. Nature 322: 73-77.
  24. Kunkel LM, Monaco AP, Middlesworth W, Ochs HD, Samuel A (1985) Specific cloning of DNA fragments absent from the DNA of a male patient with an X chromosome deletion. Proceedings of the National Academy of Sciences of the United States of America 82: 4778-4782.
  25. Takeshima Y, Yagi M, Okizuka Y, Awano H, Zhang Z, et al. (2010) Mutation spectrum of the dystrophin gene in 442 Duchenne/Becker muscular dystrophy cases from one Japanese referral center. J Human Genetics 55: 379–388.
  26. Bushby K, Finkel R, Birnkrant DJ, Case LE, Clemens PR, et al. (2010b) Diagnosis and management of Duchenne muscular dystrophy, part 2: implementation of multidisciplinary care. Lancet Neurology 9: 177–189.
  27. Hoffman EP, Dressman D (2001) Molecular pathophysiology and targeted therapeutics for muscular dystrophy. Trends Pharmacology Science 22: 465–470
  28. Nowak KJ, Davies KE (2004) Duchenne muscular dystrophy and dystrophin: Pathogenesis and opportunities for treatment. EMBO Rep 5: 872–876.
  29. Garcia-Pelagio KP, Bloch RJ, Ortega A, Gonzalez-Serratos H (2011).Biomechanics of the sarcolemma and costameres in single skeletal muscle fibers from normal and dystrophin- null mice.J Muscle Res Cell Motil 31: 323–336
  30. Muntoni F, Torelli S, Ferlini A (2003) Dystrophin and mutations: One gene, several proteins, multiple phenotypes. Lancet Neurol 2731.
  31. GenBank (2001) Homo sapiens dystrophin (DMD), transcript variant Dp427c, Mrna.  NCBI Reference Sequence: NM_000109.2
  32. GenPept (2001) Dystrophin isoform Dp427c [Homo sapiens]. NCBI Reference Sequence: NP_000100.2
  33. Michalak M, Opas M (2001). Duchenne muscular dystrophy, Chichester, John Wiley & Sons Ltd, NY, USA.
  34. Chamberlain JS., Gibbs RA, Ranier JE, Nguyen PN, Caskey CT (1988) Deletion screening of the Duchenne muscular dystrophy locus via multiplex DNA ampliï¬Âcation. Nucleic Acids Res 16: 11141–11156.
  35. Beggs AH, Koenig M, Boyce FM, Kunkel LM (1990) Detection of 98% of DMD/BMD gene deletions by polymerase chain reaction. Hum Genet 86: 45–48.
  36. Abbs S, Tuffery-Giraud S, Bakker E, Ferlini A, Sejersen T, et al. (2010) Best practice guidelines on molecular diagnostics in Duchenne/Becker muscular dystrophies. Neuromuscul Disord 20: 422.
  37. Koenig M, Beggs AH, Moyer M, Scherpf S, Heindrich K, et al. (1989) The molecular basis for Duchenne versus Becker muscular dystrophy: correlation of severity with type of deletion. Am J Hum Genet 45: 498–506.
  38. Bergen Van den JC, Wokke BH, Janson AA, Van Duinen SG, Hulsker P, et al. (2014) Dystrophin levels and clinical severity in Becker muscular dystrophy patients. J Neurol Neurosurg. Psychiatry 85: 747−753.
  39. Wilton SD, Fletcher S, Flanigan KM (2014) Dystrophin as a therapeutic biomarker: are we ignoring data from the past? Neuromuscul Disord 24: 463−466.
  40. Anthony K, Cirak S, Torelli S, Tasca G, Feng L, et al. (2011) Dystrophin quantification and clinical correlations in Becker muscular dystrophy: implications for clinical trials. Brain 134: 3547− 3559.
  41. Liechti-Gallati S, Koenig M, Kunkel LM, Frey D, Boltshauser E, et al. (1989) Molecular deletion patterns in Duchenne and Becker type muscular dystrophy. Hum Genet 81: 343−348.
  42. Dastur R, Gaitonde P, Khadilkar S, Nadkarni J (2004)  Deletion analysis of Duchenne/Becker muscular dystrophy gene using multiplex polymerase chain reaction. Ann Indian Acad Neurol 7: 333.
  43. Nishino I, Ozawa E (2002) Muscular dystrophies. Curr Opin Neurol 15: 539.
  44. Worton RG, Thompson MW (1988)  Genetics of Duchenne muscular dystrophy. Annu Rev Genet 22: 601.
  45. Aartsma-Rus A, Van Deutekom JC, Fokkema IF, Van Ommen GJ, Den Dunnen JT (2006) 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 34: 135–144.
  46. Falzarano MS, Scotton C, Passarelli C, Ferlini A (2015) Duchenne muscular dystrophy: From diagnosis to therapy. Molecules 20: 18168-18184.
  47. Wagar EA (1996) Direct hybridization and amplification applications for the diagnosis of infectious diseases. J Clin Lab Anal 10: 312–325.
  48. Winn-Deen ES (1996) Automation of molecular genetic methods. Part 2: DNA amplification techniques. J Clin Ligand Assay 19: 21–26.
  49. Rahn K, De Grandis SA, Clarke RC, Curtiss R, Gyles CL (1992) Amplification of an invA gene sequence of Salmonella Typhimurium by polymerase chain reaction as a specific method of detection of Salmonella. Mol Cell Probes 6: 271–279.
  50. Wolcott MJ (1992) Advances in nucleic acid-based detection methods. Clin Microbiol Rev 5: 370–386.
  51. Bej AK, Mahbubani MH, Atlas RM (1991) Amplification of nucleic acids by polymerase chain reaction (PCR) and other methods and applications. Crit Rev Biochem Mol Biol 26: 301–334.
  52. Demidov G, Simakova T, Vnuchkova J, Bragin A (2016) A statistical approach to detection of copy number variations in PCR-enriched targeted sequencing data. BMC Bioinformatics 17: 1272-1276.
  53. Stockley TL, Akber S, Bulgin N, Ray PN (2006) Strategy for comprehensive molecular testing for Duchenne and Becker muscular dystrophies. Genet Test 10: 229–243.
  54. Hegde MR, Chin ELH, Mulle JG, Okou DT, Warren ST, et al. (2008) Microarray-based mutation detection in the dystrophin gene. Hum Mutat 29: 1091–1099.
  55. Edwards MC, Gibbs RA (1994) Multiplex PCR: Advantages, development, and applications. PCR Methods Appl 3: S65–S75.
  56. Henegariu O, Heerema NA, Dlouhy SR, Vance GH, Vogt PH (1997) Multiplex PCR: Critical parameters and step-by-step protocol. Bio-Techniques 23: 504–511.
  57. Polz MF, Cavanaugh CM (1998) Bias in template-to-product ratios in multitemplate PCR. Appl Environ Microbiol 64: 3724–3730.
  58. Brownie J, Shawcross S, Theaker J, Whitcombe D, Ferrie R, et al. (1997) The elimination of primer-dimer accumulation in PCR. Nucleic Acids Res 25: 3235–3241.
  59. Elnifroe M, Ashshi AM, Cooper RJ, Klapper PE (2000) Multiplex PCR: Optimization and application in diagnostic virology. Clinical Microbiology Reviews 559–570.
  60. Beggs AH, Hoffman EP, Snyder JR, Arahata K, Specht L, et al. (1991) Exploring the molecular basis for variability among patients with Becker muscular dystrophy: dystrophin gene and protein studies. American Journal of Human Genetics 49: 54-67.
  61. Kunkel LM, Snyder JR, Beggs AH, Boyce FM, Feener CA (1991) Searching for dystrophin gene deletions in patients with atypical presentations. In: Lindsten J, Pettersson U (eds) Etiology of human disease at the DNA level. Raven New York, USA. p. 51-60.
  62. Ankala A, Kohn JN, Hegde A, Meka A, Ephrem CL, et al. (2012) Aberrant firing of replication origins potentially explains intragenic nonrecurrent rearrangements within genes, including the human DMD gene. Genome Res 22: 25–34.
  63. Wilton SD, Fletcher S (2008) Exon skipping and Duchenne muscular dystrophy: Hope, hype and how feasible? Neurology India 56:  254-262.
  64. Aartsma-Rus A, Straub V, Hemmings R, Haas M, Schlosser-Weber G, et al. (2017). Development of exon skipping therapies for duchenne muscular dystrophy: A critical review and a perspective on the outstanding issues. Nucleic Acid Therapeutics 27(5): 251–259.
  65. Hallwirth-Pillay KD, Bill PL, Madurai S, Mubaiwa L, Rapiti P (2007) Molecular deletion patterns in Duchenne and Becker muscular dystrophy patients from KwaZulu Natal. J Neurol Sc. 252: 1–3.
  66. Schouten JP, McElgunn CJ, Waaijer R, Zwijnenburg D, Diepvens F, et al. (2002) Relative quantification of 40 nucleic acid sequences by multiplex ligation-dependent probe amplification. Nucleic Acids Res 30: e57.
  67. Murugan S, Chandramohan A, Lakshmi BR (2010) Use of multiplex ligation-dependent probe amplification (MLPA) for Duchenne muscular dystrophy (DMD) gene mutation analysis. Indian J Med Res 132: 303-311.
  68. Verma PK, Dalal A, Mittal B, Phadke SR (2012) Utility of MLPA in mutation analysis and carrier detection for Duchenne muscular dystrophy. Indian J Hum Genet 18: 91–94.
  69. Gaudio Ddel, Yang Y, Boggs BA, Schmitt ES, Lee JA, et al. (2008) Molecular diagnosis of duchenne/becker muscular dystrophy: Enhanced detection of dystrophin gene rearrangements by oligonucleotide array-comparative genomic hybridization. Human Mutation 29: 1100-1107.
  70. Shaffer LG, Bejjani BA (2006) Medical applications of array CGH and the transformation of clinical cytogenetics. Cytogenet Genome Res 115: 303–309.
  71. Gunn SR, Robetorye RS, Mohammed MS (2007) Comparative genomic hybridization arrays in clinical pathology: Progress and challenges. Mol Diagn Ther 11: 73–77.
  72. Chaudhary AG, Al-Qahtani M, Abuzenadah A, Gari M, Al-Sofyani AA, et al. (2008) Mutation analysis in Saudi Duchenne and Becker muscular dystrophy patients using multiplex PCR. Archives Med Sci 4(1): 16-21.
  73. Basak J, Dasgupta UB, Mukherjee SC, Das SK, Senapati AK, et al. (2009) Deletional mutations of dystrophin gene and carrier detection in Eastern India. Ind J Pediatrics 7: 1007-1012.
  74. Basak J, Dasgupta UB, Banerjee TK, Senapati AK, Das SK, et al. (2006) Analysis of dystrophin gene deletions by multiplex PCR in eastern India. Neurology India 54: 310-311.
  75. Sbiti A, Kerch F, El-Sefiani A (2002) Analysis of dystrophin gene deletions by multiplex PCR in Moroccan patients. J Biomed Biotechnol 2: 158–160.
  76. Basumatary LJ, Das M, Goswami M, Kayal AK (2013) Deletion pattern in the dystrophin gene in Duchenne muscular dystrophy patients in northeast India. J Neurosci Rural Pract 4: 227-229.
  77. Tayeb MT (2010) Deletion mutations in Duchenne muscular dystrophy (DMD) in Western Saudi children. Saudi J Biological Sciences 17: 237–240.
  78. Kumari D, Mital A, Gupta M, Goyle S (2003) Deletion analysis of the dystrophin gene in Duchenne and Becker muscular dystrophy patients: use in carrier diagnosis. Neurol India 51: 223-226.
  79. Rao MV, Sindhav GM, Mehta JJ (2014) Duchenne/Becker muscular dystrophy: A report on clinical, biochemical, and genetic study in Gujarat population, India. Annals of Indian Academy of Neurology 17: 303-307.
  80. Dastur RS, Gaitonde PS, Khadilkar SV, Nadkarni JJ (2008)  Becker muscular dystrophy in Indian patients: Analysis of dystrophin gene deletion patterns. Neurology India 56: 374-378.
  81. Singh R, Kabra VM (2006) Multiplex PCR for rapid detection of exonal deletions in patients of Duchenne Muscular Dystrophy. Indian J Clinical Biochem 147-151.
  82. Kohli S, Saxena R, Thomas E, Singh J, Verma IC (2010) Gene changes in Duchenne muscular dystrophy: Comparison of multiplex PCR and multiplex ligation-dependent probe amplification techniques. Neurol India 58: 852-856.
  83. Khalap NV, Joshi VP, Ladiwalla U, Khadilkar SV, Mahajan SK (1997) A report on higher frequency of DMD gene deletion in the Indian subcontinent. Ind J Hum Genet 3: 117-120.
  84. Swaminathan B, Shubha GN, Shubha D, Murthy AR, Kumar HBK, et al. (2001) Duchenne muscular dystrophy: A clinical, histopathological and genetic study at a neurology tertiary care center in southern India 2: 1.
  85. Mallikarjuna Rao GN, Hussain T, Geeta Devi N, Jain S, Chandak GR, et al. (2003) Dystrophin gene deletions in South Indian Duchenne muscular dystrophy patients. Indian J Med Sci 57: 1-6.
  86. Banerjee M, Verma IC (1997) Are there ethnic differences in deletions in the dystrophin gene? Am J Med Genet 68: 152-157.
  87. Nouri N, Najafabadi EF, Salehi M, Hosseinzadesh M, Behnam M, et al. (2014) Evaluation of multiplex ligation dependent probe amplification analysis versus multiplex polymerase chain reaction assays in the detection of dystrophin gene rearrangements in an Iranian population subset. Advance Biomedical Research 3: 72.
  88. Lee BL, Nam SH, Lee JH, Ki CS, Lee M, et al. (2012) Genetic analysis of dystrophin gene for affected male and female carriers with Duchenne/Becker muscular dystrophy in Korea. J Korean Med Sci 27: 274–280.
  89. Hung CC, Ning SY, Lin CY, Yang CC, Lee WT, et al. (2005) Denaturing HPLC Coupled with Multiplex PCR for Rapid Detection of Large Deletions in Duchenne Muscular Dystrophy Carriers. Clinical Chemistry 51(7): 1252-1256.
  90. Haghshenas M, Akbari MT, Karizi SZ, Deilamani FK, Nafissi S, et al. (2016) Evaluation of point mutations in dystrophin gene in Iranian Duchenne and Becker muscular dystrophy patients: introducing three novel variants. J Genetics 95: 325-329.
  91. Dunnen den JT, Beggs AH (2006) Multiplex PCR for identifying DMD gene deletions. Curr Protoc Hum Genet Chapter 9: Unit 9.3
  92. Barzegar M, Habibi P, Bonyady M, Topchizadeh V, Shiva SH (2015) Exon deletion pattern in Duchene muscular dystrophy in North West of Iran. Iran J Child Neurol 9: 42-48.
  93. Lalic T, Vossen RH, Coffa J, Schouten JP, Scekic MG, et al. (2005) Deletion and duplication screening in the DMD gene using MLPA. European J Human Genetics 13: 1231–1234.
  94. Lai KKS, Loa IFM, Tong TMF, Cheng LYL, Lam STS (2006)  Detecting exon deletions and duplications of the DMD gene using Multiplex Ligation-dependent Probe Amplification (MLPA). Clinical Biochem 39: 367–372.
  95. Kerr R, Robinson C, Essop FB, Krause A (2013) Genetic testing for Duchenne/Becker muscular dystrophy in Johannesburg, South Africa. South African medical journal 103: 999-1004.
  96. Hassan MJ, Mahmood S, Ali G, Bibi N, Waheed I, et al. (2008) Intragenic deletions in the dystrophin gene in 211 Pakistani Duchenne muscular dystrophy patients. Pediatr Int Apr 50(2): 162-166.
  97. Wang J, Fan Z, Kornegay JN, Styner MA (2011)  MRI-based quantification of Duchenne muscular dystrophy in a canine model. Proc SPIE 7965: 0G1–9.
  98. Yang J, Li SY, Li YQ, Cao JiQ, Feng SW, et al. (2013) MLPA-based genotype–phenotype analysis in 1053 Chinese patients with DMD/BMD. BMC Med Genet 14: 29.
  99. Lai PS, Takeshima Y, Adachi K, Van Tran K, Nguyen HT, et al. (2002) Comparative study on deletions of the dystrophin gene in three Asian populations. Hum Genet 47: 552–555.
  100. Sura T, Eu-Ahsunthornwattana J, Pingsuthiwong S, Busabaratana M (2008) Sensitivity and frequencies of dystrophin gene mutations in Thai DMD/BMD patients as detected by multiplex PCR. Dis Markers 25: 115-121.
  101. Marini M, Salmi AA, Watihayati MS, SMardziah MD, Zahri MK, et al. (2008) Screening of dystrophin gene deletions in Malaysian patients with Duchenne muscular dystrophy. Med J Malaysia 63: 31-34.
  102. Dastur RS, Kachwala MY, Khadilkar SV, Hegde MR, Gaitonde PS (2011) Identification of deletions and duplications in the Duchenne muscular dystrophy gene and female carrier status in western India using combined methods of multiplex polymerase chain reaction and multiplex ligation-dependent probe amplification. Neurology India 59: 803-809.
  103. Singh V, Sinha S, Mishra S, Chaturvedi LS, Pradhan S, et al. (1997) Proportion and pattern of dystrophin gene deletions in north Indian Duchenne and Becker muscular dystrophy patients. Hum Genet 99: 206-208.
  104. Datkhile KD, Thorat KV, Patil MN, Vhaval RD, Mandiri VG (2015) The spectrum of dystrophin gene mutations in duchene muscular dystrophy patients of South-Western Maharashtra in India. Journal of Krishna Institute of Medical Sciences University (JKIMSU) 4: 70-80.
  105. Douglas AGL, Wood MJA (2013) Splicing therapy for neuromuscular disease. Molecular and Cellular Neuroscience 56: 169-185.
  106. Roberts RG, Gardner RJ, Bobrow M (1994) Searching for the 1 in 2,400,000: A review of dystropltain gene point mutations. Human Mutation 4: 1-11.