ArticlePDF Available

Abstract and Figures

Analysing the type and frequency of patient specific mutations that give rise to Duchenne Muscular Dystrophy (DMD) is an invaluable tool for diagnostics, basic scientific research, trial planning and improved clinical care. Locus specific databases (LSDBs) allow for the collection, organization, storage and analysis of genetic variants of disease. Here we describe the development and analysis of the TREAT-NMD DMD Global database (http://umd.be/TREAT_DMD/). We analysed genetic data for 7149 DMD mutations held within the database. 5682 large mutations were observed (80% of total mutations), of which 4894 (86%) were deletions (1 exon or larger), and 784 (14%) were duplications (1 exon or larger). There were 1445 small mutations (smaller than 1 exon, 20% of all mutations), of which 358 (25%) were small deletions and 132 (9%) small insertions and 199 (14%) affected the splice sites. Point mutations totalled 756 (52% of small mutations) with 726 (50%) nonsense mutations and 30 (2%) missense mutations. Finally, 22 (0.3%) mid-intronic mutations were observed. In addition, mutations were identified within the database that would potentially benefit from novel genetic therapies for DMD including stop codon read-through therapies (10% of total mutations) and exon skipping therapy (80% of deletions and 55% of total mutations). This article is protected by copyright. All rights reserved. This article is protected by copyright. All rights reserved.
Content may be subject to copyright.
This article has been accepted for publication and undergone full peer review but has not been through the
copyediting, typesetting, pagination and proofreading process, which may lead to differences between this
version and the Version of Record. Please cite this article as doi: 10.1002/humu.22758.
This article is protected by copyright. All rights reserved. 1
Humu-2014-0536
Databases
The TREAT-NMD DMD Global database: Analysis of more than 7000 Duchenne
Muscular Dystrophy mutations
*Catherine L. Bladen1, *David Salgado2, Soledad Monges3, Maria E. Foncuberta3, Kyriaki
Kekou4, Konstantina Kosma4, Hugh Dawkins5, Leanne Lamont5, Anna J. Roy6, Teodora
Chamova7, Velina Guergueltcheva7, Sophelia Chan8, Lawrence Korngut9, Craig Campbell10,
Yi Dai11, Jen Wang12, Nina Barišić13, Petr Brabec14, Jaana Lahdetie15, Maggie C. Walter16,
Olivia Schreiber-Katz16, Veronika Karcagi17, Marta Garami17, Venkatarman Viswanathan18,
Farhad Bayat19, Filippo Buccella20, En Kimura21, Zaïda Koeks22, Janneke C. van den
Bergen22, Miriam Rodrigues23, Richard Roxburgh23, Anna Lusakowska24, Anna Kostera-
Pruszczyk24, Janusz Zimowski25, Rosário Santos26, Elena Neagu27, Svetlana Artemieva28,
Vedrana Milic Rasic29, Dina Vojinovic29, Manuel Posada30, Clemens Bloetzer31, Pierre-Yves
Jeannet31, Franziska Joncourt32, Jordi Díaz-Manera33, Eduard Gallardo33, A. Ayşe
Karaduman34, Haluk Topaloğlu35, Rasha El Sherif36, Angela Stringer37, Andriy V. Shatillo38,
Ann S. Martin39, Holly L. Peay39, Matthew I, Bellgard40, Jan Kirschner41, Kevin M.
Flanigan42, Volker Straub1, Kate Bushby1, Jan Verschuuren22, Annemieke Aartsma-Rus1,43,
Christophe Beroud2,44, Hanns Lochmüller1.
This article is protected by copyright. All rights reserved. 2
*Equal contribution
1The John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine,
Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK. 2Aix Marseille Université, Inserm,
GMGF UMR_S 910, 13385, Marseille, France. 3Hospital de Pediatría J. P. Garrahan,
Pichincha 1881, Argentina. 4Department of Medical Genetics, Medical School, University of
Athens, Choremio Research Laboratory,St. Sophia's Children's Hospital Thinon & Levadia
Goudi, Athens, 11527, Greece. 5Office of Population Health Genomics, Department of
Health, Perth, Western Australia, Australia. 6WIV-ISP, Juliette Wytsman 14, 1050 Brussels,
Belgium. 7Department of Neurology, Medical University-Sofia, 1 Georgi Sofiiski Str, Sofia,
Bulgaria. 8Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, the
University of Hong Kong. 9Department of Clinical Neurosciences and Hotchkiss Brain
Institute, University of Calgary, Calgary, Canada; Address: Clinical Neurosciences, South
Health Campus, 4448 Front Street SE, Calgary, Alberta, Canada. 10Department of Paediatrics,
Clinical Neurological Sciences & Epidemiology, Western University, London, Ontario,
Canada. 11Department of Neurology, Peking Union Medical College Hospital, Peking Union
Medical College and Chinese Academy of Medical Sciences, Beijing, China. 12China Dolls,
Department of Neurology, the General Hospital of Chinese Armed Police Forces, 69
Yongding Road, Haidian District, Beijing 100039, P.R. China. 13Division of Paediatric
Neurology, University Hospital Centre Zagreb (KBC Zagreb), University of Zagreb Medical
School, Croatia. 14Institute for Biostatistic and Analyses, Masaryk University, Kamenice
126/3, 625 00 Brno, Czech Republic. 15Dept. Child Neurology, Turku University Central
Hospital, Turku, Finland. 16Friedrich-Baur-Institute, Department of Neurology, Ludwig-
Maximilians-University of Munich, Ziemssenstr. 1a, Munich, 80336, Germany. 17NIEH,
Dept. of Molecular Genetics and Diagnostics, Albert Florian str 2-6, Budapest, 1097,
Hungary. 18CHILDS Trust Medical Research Foundation and Apollo Children's Hospital,
Chennai, India. 19Pasteur Institute of Iran, Karaj complex, Tehran, Iran. 20Parent Project
Onlus, Via Nicola Coviello n.12, Roma, Italy. 214-1-1 Ogawa-Higashi, Kodaira,Tokyo 187-
8551, Japan. 22Leiden University Medical Center, Department of Neurology, Albinusdreef 2,
2333 ZA Leiden, The Netherlands. 23Department of Neurology, Auckland DHB, Private
Bag 92024, Auckland, New Zealand. 24Department of Neurology, Medical University of
Warsaw, Warsaw, Poland. 25Department of Genetics , Institute of Psychiatry and Neurology,
Warsaw, Poland. 26Centro de Genética Médica Jacinto Magalhães, Praça Pedro Nunes 88,
4099-028, Porto, Portugal. 27National Institute of Legal Medicine, Genetics Laboratory 9,
This article is protected by copyright. All rights reserved. 3
Vitan Barzesti, Bucharest, 042122. 28Moscow Institute of Pediatrics, 2, Taldomskaya str,
Moscow, Russia. 29Clinic for Neurology and Psychiatry for Children and Youth, Faculty of
Medicine, University of Belgrade, Dr Subotica 6A, 11000 Belgrade, Serbia. 30Institute of
Rare Diseases Research, SpainRDR and CIBERER, Institute of Health Carlos III, Madrid,
Spain. 31Neurorehabilitation Unit, Lausanne University Hospital, Lausanne, Switzerland.
32Division of Human Genetics, Children's University Hospital, Inselspital, Bern,
Switzerland.33Unitat de Malalties Neuromusculars, Servei de Neurologia, Hospital de la
Santa Creu i Sant Pau de Barcelona. 34Hacettepe University Faculty of Health Sciences
Department of Physiotherapy and Rehabilitation and and Hacettepe University Faculty of
Health Sciences Department of Physiotherapy and Rehabilitation 06100, Altındağ, Ankara,
Turkey. 35Hacettepe Children's Hospital 06100 Ankara, Turkey. 36Neurology& Neurogenic
Unit, Egypt Air Hospital, Ain Shams University, Egypt. 37Action Duchenne, Epicentre, 41
West Street, London E11 4LJ, UK. 38Institute of Neurology, Psychiatry and Narcology of
NAMS, Academic Pavlov st. 46, Kharkiv 61068, Ukraine. 39DuchenneConnect, 401
Hackensack Ave, 9th Floor, Hackensack, NJ 07601, USA. 40Centre for Comparative
Genomics, Murdoch University, Murdoch, Western Australia, 6150. 41University Medical
Center Freiburg, Germany. 42Center for Gene Therapy, The Research Institute, WA3014,
Nationwide Children‟s Hospital, 700 N. Children‟s Drive, Columbus, Ohio 43205. 43Leiden
University Medical Center, Department of Human Genetics, Leiden, The Netherlands.
44INSERM APHM, Hôpital d'Enfants de la Timone, Département de Génétique Médicale et
de Biologie Cellulaire, 13385, Marseille, France.
This article is protected by copyright. All rights reserved. 4
Corresponding Author:
Catherine L. Bladen
University of Newcastle
Institute if Genetic Medicine
Times Square
Newcastle upon Tyne
Newcastle
NE13BZ
United Kingdom
E-mail: catherine.bladen@ncl.ac.uk
Funding: This work was supported by TREAT-NMD operating grants, FP6 LSHM-CT-
2006-036825, 20123307 UNEW_FY2013 and AFM 16104. Further support came from the
European Union Seventh Framework Programme (FP7/2007-2013) under grant agreement
No. 305444 (RD-Connect) and 305121 (Neuromics).
This article is protected by copyright. All rights reserved. 5
Abstract
Analysing the type and frequency of patient specific mutations that give rise to Duchenne
Muscular Dystrophy (DMD) is an invaluable tool for diagnostics, basic scientific research,
trial planning and improved clinical care. Locus specific databases (LSDBs) allow for the
collection, organization, storage and analysis of genetic variants of disease. Here we describe
the development and analysis of the TREAT-NMD DMD Global database
(http://umd.be/TREAT_DMD/). We analysed genetic data for 7149 DMD mutations held
within the database. 5682 large mutations were observed (80% of total mutations), of which
4894 (86%) were deletions (1 exon or larger), and 784 (14%) were duplications (1 exon or
larger). There were 1445 small mutations (smaller than 1 exon, 20% of all mutations), of
which 358 (25%) were small deletions and 132 (9%) small insertions and 199 (14%) affected
the splice sites. Point mutations totalled 756 (52% of small mutations) with 726 (50%)
nonsense mutations and 30 (2%) missense mutations. Finally, 22 (0.3%) mid-intronic
mutations were observed. In addition, mutations were identified within the database that
would potentially benefit from novel genetic therapies for DMD including stop codon read-
through therapies (10% of total mutations) and exon skipping therapy (80% of deletions and
55% of total mutations).
Key Words: DMD; Duchenne muscular dystrophy; TREAT-NMD; rare disease registries
This article is protected by copyright. All rights reserved. 6
Introduction
Duchenne muscular dystrophy (DMD) is a severe, X-linked, progressive neuromuscular
disease caused by mutations in the DMD gene (DMD; MIM# 310200) (Hoffman, et al., 1988).
Mutations in this gene give rise to two forms of muscular dystrophy depending on whether
the translational reading frame is lost or maintained: severe DMD, due to out of frame
mutations leading to loss of protein function, or a milder form of muscular dystrophy known
as Becker muscular dystrophy (BMD; MIM# 300376), caused by a reduction in the amount
and/or size of dystrophin protein due to frame maintaining mutations (Koenig, et al., 1989).
The DMD gene is the largest known gene in humans, spanning 2.3 Mb of genomic DNA. The
coding sequence spans 11 Kb and is made up of 79 exons (Ahn and Kunkel, 1993). Many
different types of mutation have been described for DMD including large deletions and
duplications, point mutations and small rearrangements.
DMD has a prevalence of 21.2/100,000 school aged boys (Mah, et al., 2014). Current care
recommendations (specifically, the use of corticosteroids, cardiac medications, and assisted
ventilation) improve outcomes and quality of life but do not modify the underlying
progression of the disease (Hoffman, et al., 2012; Sejerson, et al., 2009). Potential treatment
strategies centre primarily on targeted mitigation of the causative genetic mutation. One
example of a genetic based potential therapy is non-sense stop codon read-through therapy
(Hirawat, et al., 2007; Howard, et al., 2000; Wagner, et al., 2001; Welch, et al., 2007). These
treatments, include aminoglycosides and ataluren (previously PTC124), and work by
selectively inducing ribosomal read-through of premature stop codons but not normal stop
codons. Specific non-sense mutations exist in DMD patients leading to premature stop
codons (TGA, TAG and TAA) and would potentially benefit from this therapy. Translarna
recently obtained conditional marketing authorization from the European Medicine Agency
(EMA) for use in ambulant Duchenne patients over 5 years of age, and as such is the first
drug to be approved for DMD.
This article is protected by copyright. All rights reserved. 7
A further example is the exon skipping approach. Exon skipping aims to moderate disease
progression by taking advantage of the knowledge that internally deleted dystrophins (seen in
BMD) can be partially functional (Aartsma-Rus, et al., 2009; Beroud, et al., 2007; van
Ommen and Aartsma-Rus, 2013). Significant research has been undertaken in the field of
exon skipping to restore the open reading frame of dystrophin transcripts resulting in the
production of partly functional dystrophin protein (Aartsma-Rus, et al., 2009; van Ommen, et
al., 2008). Exon skipping is achieved by the use of antisense oligonucleotides (AONs) that
specifically bind to and hide exons from the splicing machinery, leading to an in-frame
mRNA without this exon and giving rise to internally deleted dystrophin proteins as seen in
BMD patients (Aartsma-Rus, et al., 2003; Aartsma-Rus, et al., 2004; Arechavala-Gomeza, et
al., 2007; Gurvich, et al., 2008; McClorey, et al., 2006; Surono, et al., 2004; Takeshima, et al.,
2001). Since DMD has a relatively high rate of new mutations (1 in 3 mutations is new), most
patients have unique mutations (Aartsma-Rus, et al., 2006; Tuffery-Giraud, et al., 2009).
However, approximately 60%-65% of all DMD patients carry a deletion of one or more
exons, with a tendency to cluster between exons 45 and 55 (Aartsma-Rus, et al., 2006;
Tuffery-Giraud, et al., 2009). Furthermore, while the location of the breakpoints in introns
will differ for patients with (e.g.) a deletion of exon 48-50, these deletions will give rise to
identical transcripts. Therefore, the skipping of certain exons would be applied to relatively
large numbers of patients.
Understanding the type and frequency of patient specific mutations that give rise to DMD
associated phenotypes is an invaluable tool both for genetic diagnosis, basic scientific
research and improved clinical care, potentially leading to new treatments for the disease.
Currently the TREAT-NMD DMD Global database contains over 7000 (7149 as of
November 2013) mutations (http://umd.be/TREAT_DMD/). Locus specific databases
(LSDBs) allow for the collection, organization, storage and analysis of genetic variants of
This article is protected by copyright. All rights reserved. 8
disease. LSDBs collect all published and unpublished mutations for a specific gene along
with complete clinical and phenotypic information. Additional confidence exists in these
datasets due in part to the role of experts or “curators”. Curators validate the data held within
the database and significantly reduce error rates (Beroud, et al., 2005; Cotton, et al., 2008). In
the case of LSDBs for DMD, a number of databases exist including, the Leiden muscular
dystrophy pages, (http://www.dmd.nl/) in the Netherlands (Aartsma-Rus, et al., 2006), and
the UMD-DMD, (http://www.umd.be/DMD/) in France (Cotton, et al., 2008). We here report
a new global mutation database for DMD, and outline how this can be used for genetic
analysis and development of genetic therapies.
Methods
A new global database for DMD (TREAT-NMD DMD Global database) based on the French
UMD-DMD system has been developed with TREAT-NMD collaboration. TREAT-NMD
was initially established as an EU funded „network of excellence‟ with the remit of
„reshaping the research environment‟ in the neuromuscular field ([http://www.treat-nmd.eu/],
2013; Bushby, et al., 2009). Standardised mutation (DMD mutations) specific data based on
TREAT-NMD mandatory and highly encouraged items from the national TREAT-NMD
DMD registries (Bladen, et al., 2013) were transferred to the global DMD database via a
secure File Transfer Protocol (FTP) transfer in November 2013, in order to provide a single
cohort of genetic and clinical variants (Figure 1). Analysis of DMD genetic mutations was
then carried out for the 7149 patient data sets held within the TREAT-NMD DMD Global
database. HGVS nomenclature was used throughout (http://www.hgvs.org/mutnomen/).
This article is protected by copyright. All rights reserved. 9
Results
The TREAT-NMD DMD Global database currently contains 7149 Duchenne Muscular
Dystrophy (DMD) mutations. There were 5684 large mutations (80%), of which 4894 (86%)
were deletions (1 exon or larger), and 784 (14%) were duplications (1 exon or larger) (Table
1). There were 1445 small mutations (20% of all mutations), of which 358 (25%) were
deletions (smaller than 1 exon) and 132 (9%) were duplications (smaller than one exon); 199
(14%) splice site mutations were recorded. Point mutations totalled 756 (10% of all mutations,
52% of small mutations) with 726 nonsense mutations (10% of all mutations, 50% of small
mutations) and 3 missense mutations (<1% of all mutations, 2% of small mutations). Finally
22 (less than 1% of all mutations, 0.3% of small mutations) mid-intronic mutations were
observed (Table 1).
Large Mutations
Large deletions
4894 large deletions were reported and accounted for 68% of total mutations. Figure 2a,
highlights the ten most commonly reported (observed more than 100 times) large deletions
within the database. The most common large deletion was a deletion of exon 45, which was
recorded 316 times in the database (4% of deletions). Distribution of large deletions was
non-random with the majority of large deletions (80%) covering either the distal region
mutation hot spot of the dystrophin gene (exons 45-55) or the proximal region (exons 2-20)
(Figure 3).
This article is protected by copyright. All rights reserved. 10
Large duplications
784 large duplications were reported and accounted for 11% of total mutations. Figure 2b,
shows the eight most commonly reported large duplications (observed more than 10 times).
The most common large duplication was duplication of exon 2 (11% of duplications).
Distribution of large duplications was non-random with most large duplications involving
distal or proximal mutation hot spots (65%) (Figure 3).
Single exon deletions and duplications
The five most frequent single exon deletions recorded in the database (all reported more than
100 times) were, deletion of exon 45 (4%), 51 (3%), 44 (3%), 52 (3%) and 50 (2%). Single
exon duplications occurred less frequently than single exon deletions. Duplication of exon 2
was the most frequent and was reported 50 times, while the second most frequent single exon
duplication was duplication of exon 17 and was reported 11 times in the database.
Small Mutations
The database contained 1445 small lesions and included deletions (smaller than 1 exon; 355,
25%), duplications (smaller than one exon; 132, 9%), 199 (14%) splice site mutations and
756 (52%) point mutations, 726 (50%) nonsense mutations, 30 (2%) missense mutations and.
22 (0.3%) mid-intronic mutations (Table 1). Small deletions and mutations ranged in size
from two nucleotides (occurring 85 times) to 111 nucleotides (occurring twice).
This article is protected by copyright. All rights reserved. 11
Nonsense mutations
Non-sense mutations represented 50% of the small mutations in the database and 10% of total
mutations. Transition mutational events (70%) were more common than transversions (30%),
with the C-to-T substitution being the most frequent (90%).
Potential DMD Therapies
Non-sense stop codon read-through therapy has obtained conditional marketing authorization
(Hirawat, et al., 2007; Howard, et al., 2000; Wagner, et al., 2001; Welch, et al., 2007). This
treatment selectively induces ribosomal read-through of premature stop codons but not
normal stop codons. Mutations were identified within the database that would potentially
benefit from this therapy. These included 317 mutations (4% of overall mutations) with a
premature TGA stop codon, 215 (3%) with a TAG stop codon and 194 (3%) with a TAA stop
codon.
Exon skipping technology takes advantage of the fact that internally deleted dystrophins,
often seen in BMD, can be partially functional. Mutations were identified within the database
that would potentially benefit from exon skipping therapy. The top ten exon skips which
would be applicable to the largest group of patients were skipping of exon 51 (14% of total
mutations/21% of deletions), 53 (10%/15%), 45 (9%/13%), 44 (7%/11%),43 (7%/11%),
46(5%/7%), 50(4%/6%), 52(4%/5%), 55(3%/4%) and 8(2%/3%) respectively shown in Table
2. It is important to point out that the applicability of exon skipping of certain exons reduces
once antisense oligonucleotides targeting other exons have been developed. For example, the
reading frame of exon 52 deletions can be restored by skipping exon 51 or by skipping exon
53. However, once an antisense oligonucleotide for exon 51 has been developed and
approved for clinical use, the additional applicability of exon 53 skipping is then lower than
the a priori applicability, since it now only applies to 8% of mutations rather than 10%,
because the exon 52 deletion has already been rescued by exon 51 skipping. The top 10 of
exon skips and their added applicability (i.e. taking this adjustment into account) is shown in
(Table 2).
This article is protected by copyright. All rights reserved. 12
CpG sites
Substitutions involving a CpG dinucleotide accounted for 31% (233/756) of point mutations
within the database. The CpG dinucleotide has been shown to undergo oxidative deamination
of 5-methyl cytosine resulting in a mutational “hot spot and mutation rates an order of
magnitude higher than normally expected (Akalin, et al., 1994; Flanigan, et al., 2009;
Krawczak, et al., 1998).
Geography and DMD mutations
Regardless of geographical location (determined by continent), large deletions were by far
the most commonly observed (64% in Oceania to 88% in Africa) mutation followed by large
duplications (5% in Africa to 12% in Europe). The number of small mutations was generally
more variable (7% in Africa to 22% in Oceania) but this variability is likely explained by the
fact that not all countries routinely assay for point mutations and other small lesions and
indeed numbers of patients were significantly smaller for example in Africa compared to
Europe (Figure 4.).
This article is protected by copyright. All rights reserved. 13
Discussion
Databases
Resources existed for DMD prior to the creation of the TREAT-NMD DMD Global database,
the Leiden muscular dystrophy pages, (http://www.dmd.nl/) in the Netherlands (Aartsma-Rus,
et al., 2006), and the UMD-DMD, (http://www.umd.be/DMD/) in France (Cotton, et al.,
2008). For both the previously existing databases, bias of recorded mutations has been an
inherent problem due in part to the method used to determine the mutation. Historically,
detection of deletions and duplications was easier than detection of point mutations and other
small rearrangements leading to an overrepresentation of such mutations in the literature and
indeed a possible underrepresentation of point mutations and other small rearrangements.
However, this bias is becoming less of an issue due to current diagnostic techniques that are
widely available (Flanigan, et al., 2009; Prior and Bridgeman, 2005). Also, while initially,
the most commonly occurring mutations were recorded; now there is potentially a bias
towards only novel mutations being recorded (e.g.) in the Leiden database. In addition to this,
the UMD-DMD database is specific to France and could potentially include a bias for
mutations observed with higher or lower frequencies only in France. The new TREAT-NMD
DMD Global database houses what we believe to be the single largest cohort of verified
DMD mutations in the world and was established to collect and compare and molecular
mutations found within this patient group. Mutational analysis of the database illustrates the
allelic heterogeneity of the DMD gene. Indeed, one third of all DMD mutations occur de
novo (NG, 1993).
This article is protected by copyright. All rights reserved. 14
Analysis and comparisons
Analysis of the TREAT-NMD DMD Global database revealed that large deletions were the
most prevalent genetic mutation recorded and accounted for 68% of the total mutations
analysed, deletion of exon 45 being the single most common large deletion (reported 316
times). These results are similar to both the French UMD database (Tuffery-Giraud, et al.,
2009) with 62% of the mutations being large deletions and the Leiden database (Aartsma-Rus,
et al., 2006), where 72% of the mutations are large deletions. In the Leiden database and the
TREAT-NMD DMD Global databases, deletion of exon 45 was the most common deletion,
making up 4% of the mutations in the TREAT-NMD DMD Global database and 2% of the
Leiden database. Large duplications accounted for 11% of mutations in the TREAT-NMD
DMD Global database compared to 13% in the French UMD database and 8% in the Leiden
database. The most commonly occurring large duplication was duplication of exon 2 in all
three databases. Small rearrangements accounted for 20% of the TREAT-NMD DMD Global
database which was similar to the French UMD database (26%) and the Leiden database
(20%). Point mutations and nonsense mutation were the most prevalent small rearrangements
with non-sense mutations accounting for 50% of the small rearrangements in the TREAT-
NMD DMD Global database, compared to 40% in the French UMD database and 50% in the
Leiden database. Large deletions and duplications follow a non-random distribution with 78%
of them including either the proximal or distal mutation hot spots (Aartsma-Rus, et al., 2006;
Koenig, et al., 1989; Prior and Bridgeman, 2005).
This article is protected by copyright. All rights reserved. 15
Reading frame rule
The majority of the reported (DMD) mutations in the TREAT-NMD DMD Global database
resulted in frame-shift mutations (93%). Mutations not following the reading-frame rule in
the TREAT-NMD DMD Global database accounted for 7% of total mutations compared with
4% in the UMD-DMD database and 9% in the Leiden database.
Potential DMD Therapies
Several potential novel DMD therapies exist and are focused on the mitigation of the
underlying genetic defect. The two most promising examples are non-sense read through and
exon skipping. Mutations were identified within the database that would potentially benefit
from this stop codon read-through therapy (10% of mutations). Exon skipping mutations
were identified within the database that would potentially benefit from exon skipping therapy
(55% of total mutations and 80% of deletions).
Understanding the type and frequency of patient specific mutations that give rise to DMD
associated phenotypes will potentially lead to personalized (targeted/precision) therapies.
DMD essentially serves as a paradigm for this type of treatment and ultimately could lead the
way to similar approaches in other rare diseases and indeed in more common disorders.
This article is protected by copyright. All rights reserved. 16
Acknowledgments
This work was supported by TREAT-NMD operating grants, FP6 LSHM-CT-2006-036825,
20123307 UNEW_FY2013 and AFM 16104. Further support came from the European
Union Seventh Framework Programme (FP7/2007-2013) under grant agreement No. 305444
(RD-Connect) and 305121 (Neuromics).
The authors would like to acknowledge the families of those living with Duchenne muscular
dystrophy who have been instrumental in the formation of the DMD national registries. We
also acknowledge former and current members of the TREAT-NMD office at the Institute of
Genetic Medicine in Newcastle including Stephen Lynn, Emma Heslop and Rachel
Thompson. We also extend our thanks to the members of the TREAT-NMD executive
committee: Hanns Lochmuller, Annemieke Aartsma-Rus, Anna Ambrosini, Filippo Buccella,
Kevin Flanigan, Eric Hoffman, Janbernd Kirschner, Eugenio Mercuri, Ichizo Nishino, Kathy
North, Jes Rahbek and Thomas Sejersen. We also aknowledge the members of the current
TGDOC: Jan Verschuuren (Chair), Hugh Dawkins (Chair elect), Anna Ambrosini, Svetlana
Artemieva, Alexander N. Baranov, Farhad Bayat, Christophe Béroud, Ria Broekgaarden,
Filippo Buccella, Craig Campbell, Nick Catlin, Monica Ensini, Pat Furlong, Kevin Flanigan,
Ole Gredal, Lauren Hache, Serap İnal, Jacqueline Jackson, Pierre-Yves Jeannet, Anna
Kaminska, A. Ayse Karaduman, Veronika Karcagi, En Kimura, Janbernd Kirschner, Jaana
Lähdetie, Hanns Lochmüller, Vitaliy Matyushenko, Vedrana Milic-Rasic, Violeta Mihaylova,
Marie-Christine Ouillade, Ian Murphy, Miriam Rodrigues, Rosario dos Santos, Pascale
Saugier-Veber, Inge Schwersenz, Thomas Sejersen, Rasha El Sherif, Eduardo Tizzano,
Isabela Tudorache, Sylvie Tuffery-Giraud, Jen Wang, Simon Woods, W. Ludo van der Pol,
Peter Van den Bergh, Petr Vondráček.
This article is protected by copyright. All rights reserved. 17
Conflicts of interest
Professor Hanns Lochmuller was elected chair of the TREAT-NMD Alliance in April 2012
and is the previous chair of the oversight committee.
Professor Lochmuller has a financial interest/arrangement with Pfizer, Ultragenyx and
GlaxoSmithKline (Research grant investigator).
References
Aartsma-Rus A, Fokkema I, Verschuuren J, Ginjaar I, van Deutekom J, van Ommen GJ, den Dunnen JT.
2009. Theoretic applicability of antisense-mediated exon skipping for Duchenne muscular
dystrophy mutations. Hum Mutat 30:293-9.
Aartsma-Rus A, Janson AA, Kaman WE, Bremmer-Bout M, den Dunnen JT, Baas F, van Ommen GJ,
van Deutekom JC. 2003. Therapeutic antisense-induced exon skipping in cultured muscle
cells from six different DMD patients. Hum Mol Genet 12:907-14.
Aartsma-Rus A, Janson AA, Kaman WE, Bremmer-Bout M, van Ommen GJ, den Dunnen JT, van
Deutekom JC. 2004. Antisense-induced multiexon skipping for Duchenne muscular
dystrophy makes more sense. Am J Hum Genet 74:83-92.
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 34:135-44.
Ahn AH, Kunkel LM. 1993. The structural and functional diversity of dystrophin. Nat Genet 3:283-91.
Akalin N, Zietkiewicz E, Makalowski W, Labuda D. 1994. Are CpG sites mutation hot spots in the
dystrophin gene? Hum Mol Genet 3:1425-6.
Arechavala-Gomeza V, Graham IR, Popplewell LJ, Adams AM, Aartsma-Rus A, Kinali M, Morgan JE,
van Deutekom JC, Wilton SD, Dickson G et al. 2007. Comparative analysis of antisense
oligonucleotide sequences for targeted skipping of exon 51 during dystrophin pre-mRNA
splicing in human muscle. Hum Gene Ther 18:798-810.
Beroud C, Hamroun D, Collod-Beroud G, Boileau C, Soussi T, Claustres M. 2005. UMD (Universal
Mutation Database): 2005 update. Human Mutation 26:184-191.
Beroud C, Tuffery-Giraud S, Matsuo M, Hamroun D, Humbertclaude V, Monnier N, Moizard MP,
Voelckel MA, Calemard LM, Boisseau P et al. 2007. Multiexon skipping leading to an artificial
DMD protein lacking amino acids from exons 45 through 55 could rescue up to 63% of
patients with Duchenne muscular dystrophy. Hum Mutat 28:196-202.
Bladen CL, Rafferty K, Straub V, Monges S, Moresco A, Dawkins H, Roy A, Chamova T,
Guergueltcheva V, Korngut L et al. 2013. The TREAT-NMD Duchenne Muscular Dystrophy
Registries: Conception, Design, and Utilization by Industry and Academia. Hum Mutat.
Bushby K, Lynn S, Straub T, Network T-N. 2009. Collaborating to bring new therapies to the patient--
the TREAT-NMD model. Acta Myol 28:12-5.
This article is protected by copyright. All rights reserved. 18
Cotton RGH, Auerbach AD, Beckmann JS, Blumenfeld OO, Brookes AJ, Brown AE, Carrera P, Cox DW,
Gottlieb B, Greenblatt MS et al. 2008. Recommendations for locus-speciific databases and
their curation. Human Mutation 29:2-5.
Flanigan KM, Dunn DM, von Niederhausern A, Soltanzadeh P, Gappmaier E, Howard MT, Sampson JB,
Mendell JR, Wall C, King WM et al. 2009. Mutational spectrum of DMD mutations in
dystrophinopathy patients: application of modern diagnostic techniques to a large cohort.
Hum Mutat 30:1657-66.
Gurvich OL, Tuohy TM, Howard MT, Finkel RS, Medne L, Anderson CB, Weiss RB, Wilton SD, Flanigan
KM. 2008. DMD pseudoexon mutations: splicing efficiency, phenotype, and potential
therapy. Ann Neurol 63:81-9.
Hirawat S, Welch EM, Elfring GL, Northcutt VJ, Paushkin S, Hwang S, Leonard EM, Almstead NG, Ju W,
Peltz SW et al. 2007. Safety, tolerability, and pharmacokinetics of PTC124, a
nonaminoglycoside nonsense mutation suppressor, following single- and multiple-dose
administration to healthy male and female adult volunteers. J Clin Pharmacol 47:430-44.
Hoffman EP, Fischbeck KH, Brown RH, Johnson M, Medori R, Loike JD, Harris JB, Waterston R, Brooke
M, Specht L et al. 1988. Characterization of dystrophin in muscle-biopsy specimens from
patients with Duchenne's or Becker's muscular dystrophy. N Engl J Med 318:1363-8.
Hoffman EP, Reeves E, Damsker J, Nagaraju K, McCall JM, Connor EM, Bushby K. 2012. Novel
approaches to corticosteroid treatment in Duchenne muscular dystrophy. Phys Med Rehabil
Clin N Am 23:821-8.
Howard MT, Shirts BH, Petros LM, Flanigan KM, Gesteland RF, Atkins JF. 2000. Sequence specificity
of aminoglycoside-induced stop condon readthrough: potential implications for treatment of
Duchenne muscular dystrophy. Ann Neurol 48:164-9.
Koenig M, Beggs AH, Moyer M, Scherpf S, Heindrich K, Bettecken T, Meng G, Muller CR, Lindlof M,
Kaariainen H 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.
Krawczak M, Ball EV, Cooper DN. 1998. Neighboring-nucleotide effects on the rates of germ-line
single-base-pair substitution in human genes. Am J Hum Genet 63:474-88.
Mah JK, Korngut L, Dykeman J, Day L, Pringsheim T, Jette N. 2014. A systematic review and meta-
analysis on the epidemiology of Duchenne and Becker muscular dystrophy. Neuromuscul
Disord 24:482-91.
McClorey G, Moulton HM, Iversen PL, Fletcher S, Wilton SD. 2006. Antisense oligonucleotide-
induced exon skipping restores dystrophin expression in vitro in a canine model of DMD.
Gene Ther 13:1373-81.
NG L. 1993. Molecular genetics and genetic counselling for Duchenne/Becker muscular dystrophy.
Molecular and cell biology of muscular dystrohy:37-84.
Prior TW, Bridgeman SJ. 2005. Experience and strategy for the molecular testing of Duchenne
muscular dystrophy. J Mol Diagn 7:317-26.
Sejerson T, Bushby K, Excellence T-NENo. 2009. Standards of care for Duchenne muscular dystrophy:
brief TREAT-NMD recommendations. Adv Exp Med Biol 652:13-21.
Surono A, Van Khanh T, Takeshima Y, Wada H, Yagi M, Takagi M, Koizumi M, Matsuo M. 2004.
Chimeric RNA/ethylene-bridged nucleic acids promote dystrophin expression in myocytes of
This article is protected by copyright. All rights reserved. 19
duchenne muscular dystrophy by inducing skipping of the nonsense mutation-encoding exon.
Hum Gene Ther 15:749-57.
Takeshima Y, Wada H, Yagi M, Ishikawa Y, Ishikawa Y, Minami R, Nakamura H, Matsuo M. 2001.
Oligonucleotides against a splicing enhancer sequence led to dystrophin production in
muscle cells from a Duchenne muscular dystrophy patient. Brain Dev 23:788-90.
Tuffery-Giraud S, Beroud C, Leturcq F, Yaou RB, Hamroun D, Michel-Calemard L, Moizard MP,
Bernard R, Cossee M, Boisseau P et al. 2009. Genotype-phenotype analysis in 2,405 patients
with a dystrophinopathy using the UMD-DMD database: a model of nationwide
knowledgebase. Hum Mutat 30:934-45.
van Ommen GJ, Aartsma-Rus A. 2013. Advances in therapeutic RNA-targeting. N Biotechnol 30:299-
301.
van Ommen GJ, van Deutekom J, Aartsma-Rus A. 2008. The therapeutic potential of antisense-
mediated exon skipping. Curr Opin Mol Ther 10:140-9.
Wagner KR, Hamed S, Hadley DW, Gropman AL, Burstein AH, Escolar DM, Hoffman EP, Fischbeck KH.
2001. Gentamicin treatment of Duchenne and Becker muscular dystrophy due to nonsense
mutations. Ann Neurol 49:706-11.
Welch EM, Barton ER, Zhuo J, Tomizawa Y, Friesen WJ, Trifillis P, Paushkin S, Patel M, Trotta CR,
Hwang S et al. 2007. PTC124 targets genetic disorders caused by nonsense mutations.
Nature 447:87-91.
This article is protected by copyright. All rights reserved. 20
Figure 1. Upload of data from national TREAT-NMD DMD registries to Global
database. Standardised aggregate data from the national TREAT-NMD DMD registries was
transferred to the global DMD database via a secure FTP transfer, in order to provide a single
cohort of genetic and clinical variants.
This article is protected by copyright. All rights reserved. 21
Figure 2. Most commonly reported large mutations. Most commonly reported large
deletions (recorded 100 times or more) (a) and large duplications (recorded 10 times or more)
(b) in the TREAT-NMD DMD Global database.
This article is protected by copyright. All rights reserved. 22
Figure 3. Distribution of the most common large deletion and duplication on the DMD
gene.
This article is protected by copyright. All rights reserved. 23
Figure 4. Geography and DMD mutations. Distribution of DMD mutation types stratified
by continent.
This article is protected by copyright. All rights reserved. 24
Table 1. Type and Frequency of mutations held within the TREAT-NMD DMD
Global database
TOTAL
7149
% of total mutations
Large mutations
5682
79
Large deletions (>=1 exon)
4894
68
Large duplications (>=1 exon)
784
11
Small mutations
1445
20
Small deletions (<1 exon)
358
5
Small insertions (<1 exon)
132
2
Splice sites (<10 bp from exon)
199
3
Point mutations
756
11
Nonsense
726
10
Missense
30
0.4
Mid-intronic mutations
22
0.3
This article is protected by copyright. All rights reserved. 25
Table 2. Overview of DMD exons
(a) Overview of exons for which single exon skipping would be applicable to
the largest groups of patients.
(b) Adjusted overview of applicability of single exon skipping.
... A comprehensive analysis of genetic data for 7,149 DMD mutations contained in the TREAT-NMD DMD Global database (http://umd.be/TREAT_DMD/) found that 80% of total mutations were large mutations, 69% of which were deletions and 11% duplications of one or more exons, while the remaining 20% were small mutations (Bladen et al., 2015) (Figure 1, upper panel A). Of the small mutations, 25% were small deletions, 9% small insertions, and 14% affected splice sites. ...
... Of the small mutations, 25% were small deletions, 9% small insertions, and 14% affected splice sites. Point mutations accounted for 52% of small mutations; 50% nonsense mutations and 2% missense mutations (Bladen et al., 2015). ...
... Deep intronic variants can affect pre-mRNA splicing by activating cryptic intronic acceptor or donor sites, causing PE inclusion, and by altering regulatory sequence motifs recognized by specific RNA binding proteins (Vaz-Drago et al., 2017). Although their frequency in DMD patients has been estimated to be about 0.3% (Bladen et al., 2015), this class of pathogenic variants is most likely underestimated . For the removal of long introns, such as in DMD, a non-canonical mechanism of recursive splicing, first described in Drosophila (Burnette et al., 2005), has recently been observed in the dystrophin pre-mRNA (Gazzoli et al., 2016). ...
Article
Full-text available
Duchenne muscular dystrophy (DMD) is a severe genetic disorder characterized by progressive muscle degeneration, with respiratory and cardiac complications, caused by mutations in the DMD gene, encoding the protein dystrophin. Various DMD mutations result in different phenotypes and disease severity. Understanding genotype/phenotype correlations is essential to optimize clinical care, as mutation-specific therapies and innovative therapeutic approaches are becoming available. Disease modifier genes, trans-active variants influencing disease severity and phenotypic expressivity, may modulate the response to therapy, and become new therapeutic targets. Uncovering more disease modifier genes via extensive genomic mapping studies offers the potential to fine-tune prognostic assessments for individuals with DMD. This review provides insights into genotype/phenotype correlations and the influence of modifier genes in DMD.
... DMD is caused by mutations of the dystrophin gene on the X-chromosome and therefore mainly affects boys. About two thirds of patients have larger mutations with deletion or duplication of several exons, while the remaining patients harbor point mutations including premature stop codon (nonsense) and splice site mutations [3]. While DMD is typically associated with almost complete loss of dystrophin expression, Becker muscular dystrophy (BMD) is an allelic disorder caused by hypomorphic mutations associated with some residual dystrophin expression and a milder phenotype. ...
Article
Full-text available
Objective: The objective of this study was to describe predictors of loss of ambulation in Duchenne muscular dystrophy (DMD). Methods: This systematic review and meta-analysis included searches of MEDLINE ALL, Embase, and the Cochrane Database of Systematic Reviews from January 1, 2000, to December 31, 2022, for predictors of loss of ambulation in DMD. Search terms included "Duchenne muscular dystrophy" as a Medical Subject Heading or free text term, in combination with variations of the term "predictor". Risk of bias was assessed using the Newcastle-Ottawa Scale. We performed meta-analysis pooling of hazard ratios of the effects of glucocorticoids (vs. no glucocorticoid therapy) by fitting a common-effect inverse-variance model. Results: The bibliographic searches resulted in the inclusion of 45 studies of children and adults with DMD from 17 countries across Europe, Asia, and North America. Glucocorticoid therapy was associated with delayed loss of ambulation (overall meta-analysis HR deflazacort/prednisone/prednisolone: 0.44 [95% CI: 0.40-0.48]) (n = 25 studies). Earlier onset of first signs or symptoms, earlier loss of developmental milestones, lower baseline 6MWT (i.e.,<350 vs. ≥350 metres and <330 vs. ≥330 metres), and lower baseline NSAA were associated with earlier loss of ambulation (n = 5 studies). Deletion of exons 3-7, proximal mutations (upstream intron 44), single exon 45 deletions, and mutations amenable of skipping exon 8, exon 44, and exon 53, were associated with prolonged ambulation; distal mutations (intron 44 and downstream), deletion of exons 49-50, and mutations amenable of skipping exon 45, and exon 51 were associated with earlier loss of ambulation (n = 13 studies). Specific single-nucleotide polymorphisms in CD40 gene rs1883832, LTBP4 gene rs10880, SPP1 gene rs2835709 and rs11730582, and TCTEX1D1 gene rs1060575 (n = 7 studies), as well as race/ethnicity and level of family/patient deprivation (n = 3 studies), were associated with loss of ambulation. Treatment with ataluren (n = 2 studies) and eteplirsen (n = 3 studies) were associated with prolonged ambulation. Magnetic resonance biomarkers (MRI and MRS) were identified as significant predictors of loss of ambulation (n = 6 studies). In total, 33% of studies exhibited some risk of bias. Conclusion: Our synthesis of predictors of loss of ambulation in DMD contributes to the understanding the natural history of disease and informs the design of new trials of novel therapies targeting this heavily burdened patient population.
... Наиболее распространенными мутациями при МДД являются делеции (~65 %) и нонсенс-мутации (~13 %). Точечные и малые мутации, а также дупликации составляют ~12 и 10 % от общего числа мутаций соответственно [6]. Заболеваемость МДД оценивается как 1 случай на 3500-6000 новорожденных мальчиков [5]. ...
Article
Background . Duchenne muscular dystrophy (DMD) is a severe genetic disease that usually affects boys and it is characterized by a gradual loss of muscle strength up to respiratory arrest from the childhood. Currently, there are several types of successful pathogenic therapies for the disease, but it is most effective before the age of 5 years. Thereby, the problem of verifying the diagnosis before the treatment fails to work (when treatment can still make the patient’s life easier) becomes urgent. In the Russian Federation only about 1,500 boys are diagnosed with DMD, when the calculated value is 3,500. Aim . To identify all cases of DMD among patients in the neurological departments of hospitals in the Krasnoyarsk region by measuring the level of creatine phosphokinase. Materials and methods . This study was estimated by neurologists in the Krasnoyarsk Interdistrict Children’s Clinical Hospital No. 1 and the Krasnoyarsk Regional Clinical Center for Maternal and Child Health. When elevated levels of creatine phosphokinase were detected in children, genetic analysis was performed to verify DMD. Results and conclusion . Innovate experience of Krasnoyarsk region made it possible to identify all patients with DMD in the neurological departments of the Krasnoyarsk Interdistrict Children’s Clinical Hospital No. 1 and the Krasnoyarsk Regional Clinical Center for Maternal and Child Health using cheap creatine phosphokinase level analysis. The number of patients diagnosed with DMD is now ~4 cases per year. As a result, there is a correspondence between the number of real patients and the epidemiological estimate quantity for the Krasnoyarsk region.
... The higher FAIR compliance in databases focused on neuromuscular disorders could be attributed to the relatively specialized nature of these databases and patient advocacy which facilitated more focused and standardized data management practices [37,38]. Additionally, recent initiatives specially designed for data on neuromuscular disorders might have contributed to the higher FAIR adherence in these databases [39,40]. The influence of EU policies, efforts, and funding assistance that promote data sharing and FAIR implementation may be related to the observed FAIR adherence in databases with a European focus [41,42]. ...
Article
Full-text available
Background Patient registries and databases are essential tools for advancing clinical research in the area of rare diseases, as well as for enhancing patient care and healthcare planning. The primary aim of this study is a landscape analysis of available European data sources amenable to machine learning (ML) and their usability for Rare Diseases screening, in terms of findable, accessible, interoperable, reusable(FAIR), legal, and business considerations. Second, recommendations will be proposed to provide a better understanding of the health data ecosystem. Methods In the period of March 2022 to December 2022, a cross-sectional study using a semi-structured questionnaire was conducted among potential respondents, identified as main contact person of a health-related databases. The design of the self-completed questionnaire survey instrument was based on information drawn from relevant scientific publications, quantitative and qualitative research, and scoping review on challenges in mapping European rare disease (RD) databases. To determine database characteristics associated with the adherence to the FAIR principles, legal and business aspects of database management Bayesian models were fitted. Results In total, 330 unique replies were processed and analyzed, reflecting the same number of distinct databases (no duplicates included). In terms of geographical scope, we observed 24.2% ( n = 80) national, 10.0% ( n = 33) regional, 8.8% ( n = 29) European, and 5.5% ( n = 18) international registries coordinated in Europe. Over 80.0% ( n = 269) of the databases were still active, with approximately 60.0% ( n = 191) established after the year 2000 and 71.0% last collected new data in 2022. Regarding their geographical scope, European registries were associated with the highest overall FAIR adherence, while registries with regional and “other” geographical scope were ranked at the bottom of the list with the lowest proportion. Responders’ willingness to share data as a contribution to the goals of the Screen4Care project was evaluated at the end of the survey. This question was completed by 108 respondents; however, only 18 of them (16.7%) expressed a direct willingness to contribute to the project by sharing their databases. Among them, an equal split between pro-bono and paid services was observed. Conclusions The most important results of our study demonstrate not enough sufficient FAIR principles adherence and low willingness of the EU health databases to share patient information, combined with some legislation incapacities, resulting in barriers to the secondary use of data.
Article
Full-text available
In recent years, clustered regularly interspaced short palindromic repeats (CRISPRs) and CRISPR-associated (Cas) protein have emerged as a revolutionary gene editing tool to treat inherited disorders affecting different organ systems, such as blood and muscles. Both hematological and neuromuscular genetic disorders benefit from genome editing approaches but face different challenges in their clinical translation. The ability of CRISPR/Cas9 technologies to modify hematopoietic stem cells ex vivo has greatly accelerated the development of genetic therapies for blood disorders. In the last decade, many clinical trials were initiated and are now delivering encouraging results. The recent FDA approval of Casgevy, the first CRISPR/Cas9-based drug for severe sickle cell disease and transfusion-dependent β-thalassemia, represents a significant milestone in the field and highlights the great potential of this technology. Similar preclinical efforts are currently expanding CRISPR therapies to other hematologic disorders such as primary immunodeficiencies. In the neuromuscular field, the versatility of CRISPR/Cas9 has been instrumental for the generation of new cellular and animal models of Duchenne muscular dystrophy (DMD), offering innovative platforms to speed up preclinical development of therapeutic solutions. Several corrective interventions have been proposed to genetically restore dystrophin production using the CRISPR toolbox and have demonstrated promising results in different DMD animal models. Although these advances represent a significant step forward to the clinical translation of CRISPR/Cas9 therapies to DMD, there are still many hurdles to overcome, such as in vivo delivery methods associated with high viral vector doses, together with safety and immunological concerns. Collectively, the results obtained in the hematological and neuromuscular fields emphasize the transformative impact of CRISPR/Cas9 for patients affected by these debilitating conditions. As each field suffers from different and specific challenges, the clinical translation of CRISPR therapies may progress differentially depending on the genetic disorder. Ongoing investigations and clinical trials will address risks and limitations of these therapies, including long-term efficacy, potential genotoxicity, and adverse immune reactions. This review provides insights into the diverse applications of CRISPR-based technologies in both preclinical and clinical settings for monogenic blood disorders and muscular dystrophy and compare advances in both fields while highlighting current trends, difficulties, and challenges to overcome.
Article
The dystrophinopathies encompass the phenotypically variable forms of muscular dystrophy caused by pathogenic variants in the DMD gene. The dystrophinopathies include the most common inherited muscular dystrophy among 46,XY individuals, Duchenne muscular dystrophy, as well as Becker muscular dystrophy and other less common phenotypic variants. With increased access to and utilization of genetic testing in the diagnostic and carrier setting, genetic counselors and clinicians in diverse specialty areas may care for individuals with and carriers of dystrophinopathy. This practice resource was developed as a tool for genetic counselors and other health care professionals to support counseling regarding dystrophinopathies, including diagnosis, health risks and management, psychosocial needs, reproductive options, clinical trials, and treatment. Genetic testing efforts have enabled genotype/phenotype correlation in the dystrophinopathies, but have also revealed unexpected findings, further complicating genetic counseling for this group of conditions. Additionally, the therapeutic landscape for dystrophinopathies has dramatically changed with several FDA‐approved therapeutics, an expansive research pathway, and numerous clinical trials. Genotype–phenotype correlations are especially complex and genetic counselors' unique skill sets are useful in exploring and explaining this to families. Given the recent advances in diagnostic testing and therapeutics related to dystrophinopathies, this practice resource is a timely update for genetic counselors and other healthcare professionals involved in the diagnosis and care of individuals with dystrophinopathies.
Article
Full-text available
Neuromuscular diseases encompass a heterogeneous array of disorders characterized by varying onset ages, clinical presentations, severity, and progression. While these conditions can stem from acquired or inherited causes, this review specifically focuses on disorders arising from genetic abnormalities, excluding metabolic conditions. The pathogenic defect may primarily affect the anterior horn cells, the axonal or myelin component of peripheral nerves, the neuromuscular junction, or skeletal and/or cardiac muscles. While inherited neuromuscular disorders have been historically deemed not treatable, the advent of gene-based and molecular therapies is reshaping the treatment landscape for this group of condition. With the caveat that many products still fail to translate the positive results obtained in pre-clinical models to humans, both the technological development (e.g., implementation of tissue-specific vectors) as well as advances on the knowledge of pathogenetic mechanisms form a collective foundation for potentially curative approaches to these debilitating conditions. This review delineates the current panorama of therapies targeting the most prevalent forms of inherited neuromuscular diseases, emphasizing approved treatments and those already undergoing human testing, offering insights into the state-of-the-art interventions.
Preprint
Full-text available
The development of sequence-specific precision treatments like CRISPR gene-editing therapies for Duchenne Muscular Dystrophy (DMD) requires sequence humanised animal models to enable the direct clinical translation of tested strategies. The current available integrated transgenic mouse model containing the full-length human DMD gene, Tg(DMD)72Thoen/J (hDMDTg), has been found to have two copies of the transgene per locus in a tail-to-tail orientation, which does not accurately simulate the true copy number of the DMD gene. This duplication also complicates the analysis when testing CRISPR therapy editing outcomes, as large genetic alterations and rearrangements can occur between the cut sites on the two transgenes. To address this, we performed long read nanopore sequencing on hDMDTg mice to better understand the structure of the duplicated transgenes. Following that, we performed a megabase-scale deletion of one of the transgenes by CRISPR zygotic microinjection to generate a single-copy, full-length, humanised DMD transgenic mouse model (hDMDTgSc). Functional, molecular, and histological characterisation show that the single remaining human transgene retains its function and rescues the dystrophic phenotype caused by endogenous murine Dmd knockout. Our unique hDMDTgSc mouse model can potentially be used to further generation of DMD disease models, suited for the pre-clinical assessment of sequence-specific therapies.
Chapter
Muscular dystrophy (MD) corresponds to a cluster of approximately 30–40 genetically controlled diseases, which exhibit inheritance patterns that are both dominant and recessive and can be autosomal or X-linked. These disorders are marked by gradual muscle degeneration and diminished muscle potency of variable severity depending on the stage and onset age of the disease, as well as the distribution of affected muscles. In most cases, patients ultimately lose the ability to walk, and unfortunately, no therapeutic or promising drugs have been discovered for MD to date. This chapter examines the genes and the corresponding proteins, which are responsible for the majority of these conditions, as well as various diagnostic and treatment strategies, focusing on the importance of nanotechnology-based approaches. This chapter aims to provide a comprehensive understanding of the basics, clinical symptoms, and molecular mechanisms underlying various types of MDs.
Article
Full-text available
Duchenne muscular dystrophy (DMD) is an X-linked genetic disease, caused by the absence of the dystrophin protein. While many novel therapies are under development for DMD, there is currently no cure and affected individuals are often confined to a wheelchair by their teens and die in their twenties/thirties. DMD is a rare disease (prevalence < 5/10,000). Even the largest countries do not have enough affected patients to rigorously assess novel therapies, unravel genetic complexities, and determine patient outcomes. TREAT-NMD is a worldwide network for neuromuscular diseases that provides an infrastructure to support the delivery of promising new therapies for patients. The harmonized implementation of national and ultimately, global patient registries has been central to the success of TREAT-NMD. For the DMD registries within TREAT-NMD, individual countries have chosen to collect patient information in the form of standardised patient registries to increase the overall patient population on which clinical outcomes and new technologies can be assessed. The registries comprise more than 13,500 patients from 31 different countries. Here we describe how the TREAT-NMD national patient registries for DMD were established. We look at their continued growth and assess how successful they have been at fostering collaboration between academia, patient organisations and industry. This article is protected by copyright. All rights reserved.
Article
The muscular dystrophies are a broad group of hereditary muscle diseases with variable severity. Population-based prevalence estimates have been reported but pooled estimates are not available. We performed a systematic review of worldwide population-based studies reporting muscular dystrophies prevalence and/or incidence using MEDLINE and Embase databases. The search strategy included key terms related to muscular dystrophies, incidence, prevalence and epidemiology. Two reviewers independently reviewed all abstracts, full text articles and abstracted data using standardized forms. Pooling of prevalence estimates was performed using random effect models. 1104 abstracts and 167 full text articles were reviewed. Thirty-one studies met all eligibility criteria and were included in the final analysis. The studies differed widely in their approaches to case ascertainment, resulting in significant methodological heterogeneity and varied data quality. The pooled prevalence of DMD and BMD was 4.78 (95% CI 1.94-11.81) and 1.53 (95% CI 0.26-8.94) per 100,000 males respectively. The incidence of DMD ranged from 10.71 to 27.78 per 100,000. This is the first meta-analysis of worldwide prevalence estimates for muscular dystrophies. There is a need for more epidemiological studies addressing global estimates on incidence and prevalence of muscular dystrophies, utilizing standardized diagnostic criteria as well as multiple sources of case ascertainment.
Article
Detailed early chartmaking by the British East India Company and the Royal Navy in India and present-day Bangladesh provide one of the most accurate databases available to track the evolution of a major delta front over the last 200 years. Digital databases of shoreline position and shallow bathymetry of the Ganges-Brahmaputra delta front were constructed using geo-referenced and projection-corrected early and modern charts, and using LANDSAT imagery. In contrast with earlier published studies, these databases indicate the Ganges-Brahmaputra has an actively prograding subaerial delta: an average of approximately 7.0 km'/yr of new land have accreted in the river mouth region since 1792. Digitate shoals, forming in association with accretion of elongate islands in the river mouth region, are coalescing in 8-15 m water depth to form a relatively coarse-grained lobate feature that is prograding over the muddy, subaqueous delta on the inner shelf. The morphology of shoal growth suggests the Ganges-Brahmaputra mouth has evolved eastward over the late Holocene as a series of digitate shoal-channel complexes. West of the active river mouth in historical times, the delta front is sediment starved and is undergoing retreat at rates of about 1.9 kmVyr.
Article
As a result of their ability to induce translational readthrough of stop codons, the aminoglycoside antibiotics are currently being tested for efficacy in the treatment of Duchenne muscular dystrophy patients carrying a nonsense mutation in the dystrophin gene. We have undertaken a systematic analysis of aminoglycoside-induced readthrough of each stop codon in human tissue culture cells using a dual luciferase reporter system. Significant differences in the efficiency of aminoglycoside-induced readthrough were observed, with UGA showing greater translational readthrough than UAG or UAA. Additionally, the nucleotide in the position immediately downstream from the stop codon had a significant impact on the efficiency of aminoglycoside-induced readthrough in the order C > U > A ≥ G. Our studies show that the efficiency of stop codon readthrough in the presence of aminoglycosides is inversely proportional to the efficiency of translational termination in the absence of these compounds. Using the same assay, we analyzed a 33–base pair fragment of the mouse dystrophin gene containing the mdx premature stop codon mutation UAA (A), which is also the most efficient translational terminator. The additional flanking sequences from the dystrophin gene do not significantly change the relatively low-level aminoglycoside-induced stop codon readthrough of this stop codon. The implications of these results for drug efficacy in the treatment of individual patients with Duchenne muscular dystrophy or other genetic diseases caused by nonsense mutations are discussed. Ann Neurol 2000;48:164–169
Article
This paper reviews the advances in the past decade of different applications of modulating the level and content of mRNA by antisense oligonucleotide (AON)-based exon skipping. The primary aim of such modulation is the correction of genetic defects by alteration of the resulting protein such that the dysfunction is reduced or relieved. This applocation is in several clinical phase III trails, notably for Duchenne Muscular dystrophy and earlier clinical trials are in preparation for other diseases, a.o. Spinal Muscular Atrophy. A alternative aim may be to disrupt the reading frame of dysfunctional proteins when the have a dominant negative effect and their absence may ameliorate disease. A third aim is to target mRNAs for other proteins, the engineering of which might improve or prevent the disease. A final application, which is as yet under-explored but has major promise, is the functional in vivo study of protein isoforms by modulating their relative levels by AON-based skipping of alternative exons.
Article
Although prednisone has never been formally approved for use in Duchenne muscular dystrophy (DMD) by regulatory agencies, its efficacy has been confirmed in trials dating from the 1980s. There is a strong need for optimization of both specific type of glucocorticoid (eg, prednisone, vs deflazacort or others) and the dosing regimen. Ideally an optimized regimen would maximize efficacy while minimizing side-effect profiles. A new trial, FOR-DMD, aims to address this gap in knowledge. In parallel, there has been progress in the area of "dissociative steroids," drugs that are able to better separate efficacy and side effects, providing a broader therapeutic window.
Article
Aminoglycosides have previously been shown to suppress nonsense mutations, allowing translation of full-length proteins in vitro and in animal models. In the mdx mouse, where muscular dystrophy is due to a nonsense mutation in the dystrophin gene, gentamicin suppressed truncation of the protein and ameliorated the phenotype. A subset of patients with Duchenne and Becker muscular dystrophy similarly possess a nonsense mutation, causing premature termination of dystrophin translation. Four such patients, with various stop codon sequences, were treated once daily with intravenous gentamicin at 7.5 mg/kg/day for 2 weeks. No ototoxicity or nephrotoxicity was detected. Full-length dystrophin was not detected in pre- and post-treatment muscle biopsies.
Article
Care for patients with Duchenne muscular dystrophy (DMD) is poorly standardised. There are many interventions in different systems which are known to improve outcomes in DMD but these are not uniformly applied. This leads to inequality in access to treatment, as well as problems for planning controlled trials of future therapeutics. A worldwide effort is underway to generate care guidelines for DMD, which involves the Centre for Disease Control in the USA and the TREAT-NMD network of excellence for neuromuscular diseases in Europe. In advance of the full consensus document, TREAT-NMD has worked on the generation of brief standards of care for DMD, which are presented here and are available via the TREAT-NMD website (http://www.treat-nmd.eu). Guidelines are presented for diagnostics, neurological follow up, gastrointestinal and nutritional issues, respiratory and cardiac care as well as orthopaedics, rehabilitation, psychosocial interventions and oral care.
Article
Mutations in the DMD gene, encoding the dystrophin protein, are responsible for the dystrophinopathies Duchenne Muscular Dystrophy (DMD), Becker Muscular Dystrophy (BMD), and X-linked Dilated Cardiomyopathy (XLDC). Mutation analysis has traditionally been challenging, due to the large gene size (79 exons over 2.2 Mb of genomic DNA). We report a very large aggregate data set comprised of DMD mutations detected in samples from patients enrolled in the United Dystrophinopathy Project, a multicenter research consortium, and in referral samples submitted for mutation analysis with a diagnosis of dystrophinopathy. We report 1,111 mutations in the DMD gene, including 891 mutations with associated phenotypes. These results encompass 506 point mutations (including 294 nonsense mutations) and significantly expand the number of mutations associated with the dystrophinopathies, highlighting the utility of modern diagnostic techniques. Our data supports the uniform hypermutability of CGA>TGA mutations, establishes the frequency of polymorphic muscle (Dp427m) protein isoforms and reveals unique genomic haplotypes associated with "private" mutations. We note that 60% of these patients would be predicted to benefit from skipping of a single DMD exon using antisense oligonucleotide therapy, and 62% would be predicted to benefit from an inclusive multiexonskipping approach directed toward exons 45 through 55.