Bookshelf » GeneReviews » Dystrophinopathies
 
gene
GeneReviews
PagonRoberta A
BirdThomas C
DolanCynthia R
SmithRichard JH
StephensKaren
University of Washington, Seattle2009
geneticspublic health

GeneTests Home Page About GeneTests Search GeneReviews on the GeneTests web site Laboratory Directory Clinic Directory Educational Materials Illustrated Glossary

GeneReviews provides information about selected national organizations and resources for the benefit of the reader. GeneReviews is not responsible for information provided by other organizations. Information that appears in the Resources section of a GeneReview is current as of initial posting or most recent update of the GeneReview. Search GeneTests for this disorder and select graphic element for the most up-to-date Resources information.—ED.

GeneReviews designates a molecular genetic test as clinically available only if the test is listed in the GeneTests Laboratory Directory by either a US CLIA-licensed laboratory or a non-US clinical laboratory. GeneTests does not verify laboratory-submitted information or warrant any aspect of a laboratory's licensure or performance. Clinicians must communicate directly with the laboratories to verify information.—ED.

Information in the Molecular Genetics tables is current as of initial posting or most recent update. —ED.

Genetics clinics are a source of information for individuals and families regarding the natural history, treatment, mode of inheritance, and genetic risks to other family members as well as information about available consumer-oriented resources. See the GeneTests Clinic Directory.

Support groups have been established for individuals and families to provide information, support, and contact with other affected individuals. The Resources section may include disease-specific and/or umbrella support organizations.

For current information on availability of genetic testing for disorders included in this section, see GeneTests Laboratory Directory. —ED.

Genetic counseling is the process of providing individuals and families with information on the nature, inheritance, and implications of genetic disorders to help them make informed medical and personal decisions. The following section deals with genetic risk assessment and the use of family history and genetic testing to clarify genetic status for family members. This section is not meant to address all personal, cultural, or ethical issues that individuals may face or to substitute for consultation with a genetics professional. To find a genetics or prenatal diagnosis clinic, see the GeneTests Clinic Directory.

Dystrophinopathies
[Includes: Duchenne Muscular Dystrophy (DMD), Becker Muscular Dystrophy (BMD), DMD-Associated Dilated Cardiomyopathy]

Basil T Darras, MD
Director, Neuromuscular Program
Department of Neurology
Children's Hospital
Boston
Bruce R Korf, MD, PhD,FACMG
Chair, Department of Genetics
University of Alabama-Birmingham
Birmingham
David K Urion, MD
Department of Neurology
Children's Hospital
Boston
21032008dbmd
Initial Posting: September 5, 2000.
Last Update: March 21, 2008.

*

*

*

Summary

Disease characteristics. The dystrophinopathies include a spectrum of muscle disease caused by mutations in the DMD gene, which encodes the protein dystrophin. The mild end of the spectrum includes the phenotypes of asymptomatic increase in serum concentration of creatine phosphokinase (CK) and muscle cramps with myoglobinuria and isolated quadriceps myopathy. The severe end of the spectrum includes progressive muscle diseases that are classified as Duchenne/Becker muscular dystrophy when skeletal muscle is primarily affected and as DMD-associated dilated cardiomyopathy (DCM) when the heart is primarily affected. Duchenne muscular dystrophy (DMD) usually presents in early childhood with delayed milestones, including delays in sitting and standing independently. Proximal weakness causes a waddling gait and difficulty climbing. DMD is rapidly progressive, with affected children being wheelchair bound by age 12 years. Cardiomyopathy occurs in all affected individuals after age 18 years. Few survive beyond the third decade, with respiratory complications and cardiomyopathy being common causes of death. Becker muscular dystrophy (BMD) is characterized by later-onset skeletal muscle weakness; individuals remain ambulatory into their 20s. Despite the milder skeletal muscle involvement, heart failure from DCM is a common cause of morbidity and the most common cause of death. Mean age of death is in the mid-40s. DMD-associated DCM is characterized by left ventricular dilation and congestive heart failure. Female carriers of DMD mutations are at increased risk for DCM.

Diagnosis/testing. DMD is the only gene associated with the dystropinopathies. Molecular genetic testing of DMD can establish the diagnosis of a dystrophinopathy without muscle biopsy in most individuals with DMD and BMD. Virtually all males with DMD and at least 85% of males with BMD have identifiable DMD mutations. The number of individuals with DMD-associated DCM and identifiable DMD mutations is unknown. In the remaining cases, a combination of clinical findings, family history, serum CK concentration, and muscle biopsy with dystrophin studies confirms the diagnosis.

Management. Treatment of manifestations: aggressive management of DCM with anti-congestive medications in all persons and cardiac transplantation in severe cases; prednisone to improve the strength and motor function in children with DMD unless side effects are severe; deflazacort, a synthetic derivative of prednisolone used in Europe, may have fewer side effects than prednisone; physical therapy to promote mobility and prevent contractures. Prevention of secondary complications: evaluation by pulmonologist and cardiologist before surgeries; pneumococcal and influenza immunizations annually; sunshine and a balanced diet rich in vitamin D and calcium to improve bone density and reduce the risk of fractures; weight control to avoid obesity. Surveillance: for males with DMD or BMD: annual or biannual evaluation by a cardiologist beginning around age 10 years; monitoring for scoliosis; baseline pulmonary function testing before wheelchair dependence; frequent evaluations by a pediatric pulmonologist. For carriers: cardiac evaluation at least once after the teenage years. Agents/circumstances to avoid: botulinum toxin injections. Testing of relatives at risk: identification of female carriers because of the need for cardiac surveillance. Therapies under investigation: oxandrolone, cyclosporine, aminoglycosides codons; PTC124, which promotes ribosomal read-through of nonsense (stop) mutations; stem cell therapy and gene therapy. Other: no benefit from immunosuppression with azathioprine, myoblast transfer, or creatine monohydrate.

Genetic counseling. The dystrophinopathes are inherited in an X-linked manner. The risk to the sibs of a proband depends on the carrier status of the mother. Carrier females have a 50% chance of transmitting the DMD mutation in each pregnancy. Sons who inherit the mutation will be affected; daughters who inherit the mutation are carriers and may or may not develop cardiomyopathy. Males with DMD do not reproduce. Males with BMD or DMD-associated DCM may reproduce. All of their daughters are carriers; none of the sons inherit their father's DMD mutation. Prenatal testing for pregnancies at increased risk is possible if the DMD disease-causing mutation in a family member is known or if informative linked markers have been identified.

Diagnosis

Clinical Diagnosis

In addition to a positive family history compatible with X-linked inheritance, the following clinical findings support the diagnosis of Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD), and DMD-associated dilated cardiomyopathy (DCM) in males:

Duchenne muscular dystrophy (DMD)

  • Progressive symmetrical muscular weakness, proximal greater than distal, often with calf hypertrophy

  • Symptoms present before age five years

  • Wheelchair dependency before age 13 years

Becker muscular dystrophy (BMD)

  • Progressive symmetrical muscle weakness and atrophy, proximal greater than distal, often with calf hypertrophy (weakness of quadriceps femoris may be the only sign)

  • Activity-induced cramping (present in some individuals)

  • Flexion contractures of the elbows (if present, late in the course)

  • Wheelchair dependency (if present, after age 16 years)

  • Preservation of neck flexor muscle strength (differentiates BMD from DMD)

Note: The presence of fasciculations or loss of sensory modalities excludes the diagnosis of a dystrophinopathy. Individuals with an intermediate phenotype (outliers) have symptoms of intermediate severity and become wheelchair bound between ages 13 and 16 years.

DMD-associated dilated cardiomyopathy (DCM)

  • Dilated cardiomyopathy (DCM) with congestive heart failure, with males typically presenting between ages 20 and 40 years and females presenting later in life

  • Usually no clinical evidence of skeletal muscle disease; may be classified as "subclinical" BMD

  • Rapid progression to death in several years in males and slower progression over a decade or more in females [Beggs 1997]

See also Dilated Cardiomyopathy Overview.

Testing

Serum creatine phosphokinase (CK) concentration (Table 1)

Table 1. Serum Creatine Phosphokinase (CK) Concentration in the Dystrophinopathies

Phenotype% of Affected Individuals Serum CK Concentration
Males DMD 100% 1>10X normal
BMD 100% 1>5X normal
DMD-associated DCM Most individuals 2"Increased"
Female Carriers DMD ~50% 3, 42-10X normal
BMD ~30% 3, 42-10X normal

1. Serum CK concentration gradually decreases with advancing age as a result of the progressive elimination of dystrophic muscle fibers that are the source of the elevated serum CK concentration [Hoffman et al 1988, Zatz et al 1991].

2. Serum CK concentrations are usually increased, but normal concentrations have been reported in DMD-associated DCM [Mestroni et al 1999].

4. Other investigations have confirmed a wide variability in serum CK concentration among DMD/BMD carriers with the mean serum CK concentration significantly increased in carriers younger than age 20 years compared with those older than 20 [Sumita et al 1998].

Electromyography (EMG) is useful in distinguishing a myopathic process from a neurogenic disorder. This is done by demonstrating short-duration, low-amplitude, polyphasic, rapidly recruited motor unit potentials. As the disease progresses, the interference pattern becomes incomplete because of reduced recruitment and eventually the muscle becomes electrically silent. However, these findings are nonspecific, occurring in all myogenic disorders. In practice, EMG is used only rarely in the diagnosis of dystrophinopathies.

Skeletal muscle biopsy

Histology. Muscle histology early in the disease shows nonspecific dystrophic changes, including variation in fiber size, foci of necrosis and regeneration, hyalinization, and, later in the disease, deposition of fat and connective tissue.

Western blot and immunohistochemistry are summarized in Table 2.

Table 2. Findings in the Dystrophin Protein from Skeletal Muscle Biopsy

Phenotype Western Blot Immuno- histochemistry 1
Dystrophin Molecular Weight 2Dystrophin Quantity 3
Males DMD Nondetectable 0%-5% Complete/almost complete absence
Intermediate Normal/abnormal 5%-20%
BMD Normal Abnormal 20%-50% 20%-100%Normal appearing or reduced intensity ± patchy staining
Female Carriers DMD random XCI 4Normal/abnormal >60% 5 (70±9%, Pegoraro et al [1995]) Mosaic pattern
DMD skewed XCINormal/abnormal <30% on average (29±25%, Pegoraro et al [1995]) 6Mosaic pattern

1. Uses monoclonal antibodies to the C terminus, N terminus, and rod domain of dystrophin [Hoffman et al 1988]

2. Normal molecular mass is 427 kb.

3. The quantity of dystrophin is expressed in percent of control values. The reference ranges shown in this table are the ones currently used by clinical laboratories and reflect approximate and reconciled data from the literature.

5. Quantitative analysis of dystrophin in female carriers is not useful in clinical practice because of the wide range of values and the significant overlap with normal values.

6. Intermediate, severe cases

Cytogenetic analysis

Males. In rare instances, boys with DMD have other X-linked disorders including retinitis pigmentosa, chronic granulomatous disease, and McLeod red cell phenotype (see McLeod neuroacanthocytosis syndrome) [Francke et al 1985] or glycerol kinase deficiency and adrenal hypoplasia [Darras & Francke 1988] as part of contiguous gene deletion syndromes. Such boys warrant both high-resolution chromosome studies to look for visible cytogenetic deletions or rearrangements involving Xp21.2 and FISH analysis with probes (covering the GK and NRDB1 genes in addition to exons in the DMD gene).

Females. Girls with classic DMD may have an X-chromosome rearrangement or deletion involving Xp21.2, complete absence of an X chromosome (i.e., Turner syndrome), or uniparental disomy of the X chromosome. Girls with findings of typical DMD warrant high-resolution chromosome studies.

Molecular Genetic Testing

GeneReviews designates a molecular genetic test as clinically available only if the test is listed in the GeneTests Laboratory Directory by either a US CLIA-licensed laboratory or a non-US clinical laboratory. GeneTests does not verify laboratory-submitted information or warrant any aspect of a laboratory's licensure or performance. Clinicians must communicate directly with the laboratories to verify information.—ED.

Gene. DMD is the only gene known to be associated with DMD, BMD, and DMD-associated DCM.

Clinical testing

Table 3 summarizes molecular genetic testing for this disorder.

Table 3. Molecular Genetic Testing Used in the Dystrophinopathies

Test MethodMutations
Detected
Mutation Detection Frequency in Males by Phenotype and Test MethodTest Availability
DMD BMD XLDCM
Deletion/duplication analysisDeletion of one or more exons of DMD gene~65% ~85% UnknownClinical graphic element
Duplication of one or more exons of DMD ~7%-10%~6%-10%Unknown
Mutation scanning and/or sequence analysisSmall insertions/deletions/point mutations/splicing mutations of DMD gene ~25%-30%~5%-10% Unknown

Interpretation of test results. For issues to consider in interpretation of sequence analysis results, click here.

Testing Strategy

Establishing the diagnosis of a dystrophinopathy. For individuals with clinical findings suggesting a dystrophinopathy and an elevated serum CK concentration, the first step in diagnosis is molecular genetic testing of the DMD gene:

Identifying the origin of a de novo mutation. In families with a dystrophinopathy in one family member only, the origin of a de novo mutation can often be identified by performing deletion/duplication analysis or sequencing in conjunction with linkage analysis. The haplotype associated with the mutated DMD allele in the affected individual can be tracked back through the mother and, if necessary, through maternal grandparents to identify the individual in whom the mutation originated [Ferreiro et al 2004].

Carrier testing for at-risk relatives requires prior identification of the disease-causing mutations in the family.

Note: Carriers are heterozygotes for this X-linked disorder and may develop clinical findings related to the disorder.

Prenatal diagnosis and preimplantation diagnosis for at-risk pregnancies require prior identification of the disease-causing mutation in the family.

Clinical Description

Natural History

Males

The dystrophinopathies cover a spectrum of muscle disease that ranges from mild to severe. The mild end of the spectrum includes the phenotypes of asymptomatic increase in serum concentration of CK, muscle cramps with myoglobinuria, and isolated quadriceps myopathy. The severe end of the spectrum includes progressive muscle diseases that are classified as Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) when skeletal muscle is primarily affected and as DMD-associated dilated cardiomyopathy (DCM) when the heart is primarily affected [Beggs 1997, Cox & Kunkel 1997].

The distinction between DMD and BMD is based on the age of wheelchair dependency: before age 13 years in DMD and after age 16 years in BMD. An intermediate group of individuals who become wheelchair bound between ages 13 and 16 years is also recognized. Additionally, some investigators have extended the mild end of the BMD spectrum to include individuals with elevated serum CK concentration and abnormal dystrophin on muscle biopsy, but with "subclinical" skeletal muscle involvement [Melacini et al 1996]. When these individuals with atypical disease develop severe cardiomyopathy, it is not possible to distinguish between BMD and DMD-associated DCM [Cox & Kunkel 1997].

Cardiac involvement is usually asymptomatic in the early stages of the disease, although sinus tachycardia and various ECG abnormalities may be noted. Echocardiography is normal or shows only regional abnormalities.

Subclinical or clinical cardiac involvement is present in approximately 90% of individuals with DMD/BMD; however, cardiac involvement is the cause of death in only 20% of individuals with DMD and 50% of those with BMD. DCM generally presents with congestive heart failure secondary to an increase in ventricular size and impairment of ventricular function. In males, DCM is rapidly progressive with onset in teenage years, leading to death from heart failure within one to two years after the diagnosis [Finsterer & Stollberger 2003]. Individuals with DCM may or may not have clinical evidence of skeletal muscle disease.

DMD usually presents in early childhood with delayed milestones, including delays in sitting and standing independently. The mean age of walking is approximately 18 months (range 12-24 months). The first symptoms of DMD as identified by parents are typically: general motor delays (42%); gait problems, including persistent toe-walking and flat-footedness (30%); delay in walking (20%); learning difficulties (5%); and speech problems (3%). The mean age of diagnosis of boys with DMD without a family history of DMD is approximately four years ten months (range: 16 months - 8 years) [Bushby 1999, Zalaudek et al 1999]. Proximal weakness causes a waddling gait and difficulty climbing. Boys use the Gower maneuver to rise from a supine position, using the arms to supplement weak pelvic girdle muscles. The calf muscles are hypertrophic and firm to palpation. Occasionally there is calf pain. DMD is rapidly progressive, with affected children being wheelchair bound by age 12 years.

Among children with DMD, the incidence of cardiomyopathy increases steadily in the teenage years, with approximately one-third of individuals being affected by age 14 years, one-half by age 18 years, and all individuals after age 18 years [Nigro et al 1990].

Some degree of non-progressive cognitive impairment in boys with DMD has long been known. This was initially described as a general leftward shift in the spectrum of IQ scores in the population with DMD. Earlier reports suggested that verbal IQ was more affected than performance IQ on instruments such as the Wechsler Intelligence Scales.

Hinton et al [2001] demonstrated that the verbal difficulties seen in boys with DMD were mostly the result of troubles with short-term verbal memory. More recently, Wicksell et al [2004] noted particular deficits in memory (especially active working memory) and executive function and observed that these deficits in memory occurred in visuospatial as well as verbal-auditory settings. This suggested that the verbal-performance discrepancy observed by Hinton et al was an artifact of the larger impact of active working memory on the verbal tests of the instruments used in earlier studies. Thus, a specific cognitive profile of boys with DMD has emerged, demonstrating deficits in working memory and executive function. These deficits in executive function are often confused with attention deficit /hyperactivity disorder (ADHD), particularly if questionnaires or other historic evaluation means are the only investigations used. To distinguish between ADHD and more broadly defined executive function disorders, neuropsychologic investigation is warranted.

DMD is associated with reduced mobility. Thus, boys with DMD have decreased bone density and an increased risk of fractures. Corticosteroids further increase the risk of vertebral compression fractures, many of which are asymptomatic.

Few affected individuals survive beyond the third decade. Respiratory complications and cardiomyopathy are common causes of death. Because death frequently occurs outside the hospital setting, the cause of death is often hard to determine [Parker et al 2005].

BMD is characterized by later-onset skeletal muscle weakness; individuals remain ambulatory into their 20s. Despite the milder skeletal muscle involvement, heart failure from DCM is a common cause of morbidity and the most common cause of death [Cox & Kunkel 1997]. Mean age of death is in the mid-40s [Bushby 1999]. With improved diagnostic techniques, it has been recognized that the mild end of the spectrum includes men with onset of symptoms after age 30 years who remain ambulatory even into their 60s [Yazaki et al 1999].

Mildly affected individuals with confirmatory DMD molecular genetic studies and/or dystrophin studies on muscle biopsy have been classified as having either: (1) BMD with "subclinical" skeletal muscle involvement in the presence of elevated serum CK concentration, calf hypertrophy, muscle cramps, myalgia, and exertional myoglobinuria or (2) "benign" skeletal muscle involvement when "subclinical" findings are accompanied by muscle weakness in the pelvic girdle and/or shoulder girdle [Melacini et al 1996].

Cognitive impairment is not as common or as severe as in DMD.

DMD-associated DCM. In 1987, a five-generation, 63-member family with DCM but no evidence of skeletal myopathy was reported. Males present in their teens and twenties; the disease course is rapidly progressive and associated ventricular arrythmias are common. Female carriers develop mild dilated cardiomyopathy in the fourth or fifth decade, with slow progression. The only biochemical abnormality is elevation in serum CK concentration. Towbin et al [1993] demonstrated linkage to the dystrophin locus in this family and one other. Subsequent study demonstrated that in individuals with the most severe cardiac phenotype the cardiac muscle is usually unable to produce dystrophin, while skeletal muscle is unaffected [Ferlini et al 1999].

DMD-associated DCM may be the presenting finding in individuals with BMD who have little or no clinical evidence of skeletal muscle disease. Some investigators classify such individuals as having subclinical or benign BMD, whereas others may classify such individuals having DCM with increased serum CK concentration [Towbin 1998]. In one study of 28 individuals with subclinical and benign BMD between ages six and 48 years, 19 (68%) had myocardial involvement, although only two were symptomatic [Melacini et al 1996]. In another study of 21 individuals ranging from age three to 63 years (mean age 40 years), 33% had cardiac failure despite relatively mild skeletal muscle findings [Saito et al 1996].

Females

Occasionally, females have clinical features of DMD as the result of X-chromosome rearrangements involving the DMD locus. In other instances, females who have a disease-causing DMD mutation have DMD because of Turner syndrome (i.e., complete or partial absence of an X chromosome) or non-random X-chromosome inactivation (XCI). Studies show no clear correlation between the active-to-inactive X-chromosome ratio in XCI studies in leukocytes and serum CK concentration, clinical signs, or the proportion of dystrophin-negative fibers observed on muscle biopsy [Sumita et al 1998].

In contrast, Pegoraro et al [1995] showed that more than 90% of female carriers with skewed XCI (defined as ≥75% of nuclei harboring the mutant DMD gene on the active X-chromosome) as demonstrated in blood develop moderate to severe muscular dystrophy.

Signs and symptoms of DMD and BMD were studied among confirmed carriers [Hoogerwaard et al 1999a, Hoogerwaard et al 1999b] (Table 4). In contrast, Nolan et al [2003] found no cardiac abnormalities in 23 proven carriers age 6.2 to 15.9 years.

Table 4. Signs and Symptoms in Carriers of Duchenne and Becker Muscular Dystrophy

Signs/Symptoms DMD CarriersBMD Carriers
None 76% 81%
Muscle weakness 119% 14%
Myalgia/cramps 5% 5%
Left ventricle dilation19% 16%
Dilated cardiomyopathy8% 0

1. Mild to moderate weakness

Genotype-Phenotype Correlations

In males with DMD and BMD, phenotypes are best correlated with the degree of expression of dystrophin, which is largely determined by the reading frame of the spliced message obtained from the deleted allele [Monaco et al 1988, Koenig et al 1989].

DMD. Very large deletions may lead to absence of dystrophin expression. Mutations that disrupt the reading frame include stop mutations, some splicing mutations, and deletions or duplications. They produce a severely truncated dystrophin protein molecule that is degraded, leading to the more severe DMD phenotype. Exceptions to this "reading frame rule" are deletions in protein-binding domains that may severely affect function even in in-frame [Hoffman et al 1991] and exon-skipping events in which apparently out-of-frame deletions behave as in-frame deletions or vice versa [Chelly et al 1990]. The accuracy of phenotype prediction using this rule is in the range of 91%-92% [Aartsma-Rus et al 2006b].

Data suggest that dystrophin deletions involving the brain distal isoform Dp140 are associated with intellectual impairment [Felisari et al 2000].

BMD. The BMD phenotype occurs when some dystrophin is produced, usually resulting from deletions or duplications that juxtapose in-frame exons, some splicing mutations, and most non-truncating single-base changes that result in translation of a protein product with intact N and C termini. The shorter than normal dystrophin protein molecule, which retains partial function, produces the milder BMD phenotype [Deburgrave et al 2007].

DMD-associated DCM. DMD-associated DCM is caused by mutations in DMD that affect the muscle promoter (PM) and the first exon (E1), resulting in no dystrophin transcripts being produced in cardiac muscle; however, two alternative promoters that are normally only active in the brain (PB) and Purkinje cells (PP) are active in the skeletal muscle, resulting in dystrophin expression sufficient to prevent manifestation of skeletal muscle symptoms [Beggs 1997, Towbin 1998, Yoshida et al 1998].

DMD-associated DCM may also be caused by alteration of epitopes in a region of the protein of particular functional importance to cardiac muscle [Ortiz-Lopez et al 1997] or possibly by mutations in hypothetical cardiac-specific exons. Abnormalities in cardiac conduction noted in persons with dystrophinopathies may be related to reduced expression of cardiac sodium channel NA(v)1.5 secondary to dystrophin deficiency [Gavillet et al 2006].

See also Dilated Cardiomyopathy Overview.

Penetrance

Penetrance of dystrophinopathies is complete in males.

Penetrance in carrier females varies, depending on patterns of X-chromosome inactivation.

Anticipation

Anticipation is not observed in the dystrophinopathies.

Nomenclature

The term "pseudohypertrophic muscular dystrophy" was used in the past; however, it is not used currently because pseudohypertrophy is not unique to the DMD or BMD phenotype.

Prevalence

The birth prevalence of DMD in northern England is one in 5,618 live male births, and that of BMD is one in 18,450 live male births.

Differential Diagnosis

For current information on availability of genetic testing for disorders included in this section, see GeneTests Laboratory Directory. —ED.

Limb-girdle muscular dystrophy (LGMD) is a group of disorders that are clinically similar to DMD but occur in both sexes as a result of autosomal recessive and autosomal dominant inheritance. Limb-girdle dystrophies are caused by mutations in genes that encode sarcoglycans and other proteins associated with the muscle cell membrane that interact with dystrophin [Bushby 1999]. Testing for deficiency of proteins from the transmembrane sarcoglycans complex is indicated in individuals with dystrophin-positive dystrophies. LGMD type 2I phenotypically resembles DMD/BMD and is caused by mutations in FKRP, the gene encoding fukutin-related protein [Schwartz et al 2005].

Emery-Dreifuss muscular dystrophy (EDMD) is characterized by joint contractures that begin in early childhood, slowly progressive muscle weakness and wasting initially in a humero-peroneal distribution that later extends to the scapular and pelvic girdle muscles, and cardiac involvement that may include palpitations, presyncope and syncope, poor exercise tolerance, and congestive heart failure. Age of onset, severity, and progression of the muscle and cardiac involvement show intra- and interfamilial variation. Clinical variability ranges from early and severe presentation in childhood to a late onset and slowly progressive course. In general, joint contractures appear during the first two decades, followed by muscle weakness and wasting. Cardiac involvement usually occurs after the second decade. The two genes known to be associated with EDMD are EMD, encoding emerin (X-linked EDMD), and LMNA, encoding lamins A and C (autosomal dominant EDMD and autosomal recessive EDMD).

Spinal muscular atrophy (SMA) is suspected in individuals with poor muscle tone, symmetric muscle weakness that spares the face and ocular muscles, and evidence of anterior horn cell involvement, including fasciculations of the tongue and absence of deep tendon reflexes. SMA is caused by mutations in SMN. Inheritance is autosomal recessive.

Dilated cardiomyopathy (DCM) can be familial or nonfamilial. In a large series in which family studies were performed, one-third of individuals had nonfamilial DCM and two-thirds had familial DCM. Causes of familial DCM included autosomal dominant (56%), autosomal recessive (16%), X-linked with DMD mutation (10%), autosomal dominant with subclinical muscle disease (7.7%), DCM with conduction defects (2.6%), and unclassified (7.7%) [Mestroni et al 1999].

The spectrum of X-linked infantile DCM (TAZ-related DCM), caused by mutations in TAZ, includes Barth syndrome, X-linked endocardial fibroelastosis, left ventricular non-compaction, and severe X-linked DCM [D'Adamo et al 1997, Yen et al 2008].

Management

Evaluations Following Initial Diagnosis

To establish the extent of disease in an individual diagnosed with a dystrophinopathy, the following evaluations are recommended:

  • Physical therapy assessment

  • Developmental evaluation before entering elementary school for the purpose of designing an individualized educational plan, as necessary

  • If the individual is older than age ten years at diagnosis, evaluation for cardiomyopathy by electrocardiography, chest radiography, cardiac echocardiography, pulmonary function studies, and/or MRI [Towbin 2003]

Treatment of Manifestations

Appropriate management of males with DMD/BMD can prolong survival and improve quality of life.

Dilated cardiomyopathy. Recommendations are based on an American Academy of Pediatrics policy statement [American Academy of Pediatrics Section on Cardiology and Cardiac Surgery 2005].

A retrospective observational study found that ventricular remodeling may occur in males with DMD and BMD with early diagnosis and treatment of cardiomyopathy [Jefferies et al 2005]. Among 69 affected boys with a first echocardiogram indicating dilated cardiomyopathy (e.g., LVEP <55% or left ventricular dilation), 27 with DMD and four with BMD were started on an ACE inhibitor at a mean age of 15 years. If echocardiography at three months showed no improvement, a beta blocker (carvedilol or metoprolol) was added. When these 31 individuals had repeat echocardiography at a mean of 3.3 years later, left ventricular size and function normalized in 19 (66%), improved in eight (26%), and stabilized in two (8%). Mean LVEF increased from 36% to 53%. Measurement of reduced sphericity indicated improved ventricular geometry. Whether this therapy reverses the cardiomyopathy or only masks the manifestations of a progressive process is not clear; however, a more recent study [Duboc et al 2005] lends support to the hypothesis that early introduction of afterload reduction therapy may lead to improved preservation of myocardial function [Colan 2005].

The authors' institution commonly treats children with DMD or BMD early with an ACE inhibitor and/or beta blocker. In cases of overt heart failure, other heart failure therapies including diuretics and digoxin are used as needed. Cardiac transplantation is offered to persons with severe dilated cardiomyopathy and BMD with limited or no clinical evidence of skeletal muscle disease.

  • Treatment with anti-congestive medications as needed

  • Therapeutic intervention using angiotensin-converting enzyme inhibitors and beta-blockers at the earliest signs of cardiomyopathy [Towbin 2003]

    Note: In most cases, this approach leads to normalization of left ventricular size and systolic function early on.

  • Cardiac transplantation in severe cases

Medications

Prednisone. Studies have shown that prednisone improves the strength and function of individuals with DMD. It is hypothesized that prednisone has a stabilizing effect on membranes and perhaps an anti-inflammatory effect. Whether the improvement is the result of an immunosuppressive effect remains unclear, as individuals treated with azathioprine did not have a beneficial effect.

In a randomized double-blind six-month trial, prednisone administered at a dose of either 0.75 mg/kg/day or 1.5 mg/kg/day increased strength and reduced the rate of decline in males with DMD [Mendell et al 1989]. The improvement begins within ten days of starting the treatment, requires a single dose of 0.75 mg/kg/day of prednisone for maximal improvement, reaches a plateau after three months, and can be sustained for as long as three years in those children maintained on doses of 0.5 and 0.6 mg/kg/day [Fenichel et al 1991]. One open-label study suggested that therapy with prednisone could prolong ambulation by two years. Side effects include weight gain (>20% of baseline) (40%), hypertension, behavioral changes, growth retardation, cushingoid appearance (50%), and cataracts [Mendell et al 1989, Griggs et al 1993].

Follow-up studies showed that a dose of 0.75 mg/kg/day was more beneficial than a dose of 0.3 mg/kg/day [Fenichel et al 1991]. Nevertheless, a subsequent study proved the effectiveness of lower doses (0.35 mg/kg/day) of prednisolone in both DMD and BMD. One of the authors has noted sustained effectiveness with doses initiated at 0.75 mg/kg/day (maximum daily dose: 40 mg) and gradually reduced (usually not deliberately but because of advancing age and weight gain) to as low as 0.4 mg/kg/day. At lower doses, the improvement is less robust [Darras, personal communication].

Alternate-day dosing and intermittent dosing (e.g., 10 days on, 10 days off) are also used; the optimal dosage and schedule have not been adequately studied [Dubowitz 1997]. Another study showed reduced incidence of side effects by high-dose (5 mg/kg), twice-weekly dosing [Connolly et al 2002]. A randomized, cross-over, controlled trial of intermittent prednisone (0.75 mg/kg/day) therapy (prednisone or placebo) during the first ten days of each month for six months showed that prednisone slowed deterioration of muscle function in individuals with DMD [Beenakker et al 2005]; side effects did not negatively affect quality of life. Similar conclusions regarding the effect and side-effect profile of prednisone treatment for DMD were reached by a Cochrane systematic review [Manzur et al 2004] and also by the 124th European Neuromuscular Centre workshop on the treatment of DMD [Bushby et al 2004].

Note: Long-term benefit of twice-weekly oral prednisolone (5 mg/kg) beginning at three or four weeks has also been documented in the mdx mouse model of DMD [Keeling et al 2007].

Data regarding the optimal age to begin treatment with corticosteroids or the optimal duration of such treatment are insufficient. During the past few years, it has been proposed that individuals with DMD begin treatment with low-dose prednisone as soon as the diagnosis is made (age 2-5 years) [Merlini et al 2003]. Nonetheless, the efficacy and safety of early treatment have not been studied adequately. Thus, at this point corticosteroid therapy remains the treatment of choice for affected individuals between ages five and 15 years. It should be noted that studies have suggested that an optimal window for treatment may be before age five years, but large-scale controlled trials of corticosteroid therapy in early DMD have yet to be conducted.

Immunosuppressive therapy. The following recommendations for immunosuppressive therapy are in accordance with the national practice parameters regarding corticosteroid therapy developed by the American Academy of Neurology and the Child Neurology Society [Moxley et al 2005].

  • Boys with DMD who are older than age five years should be offered treatment with prednisone (0.75/mg/kg/day). Prior to the initiation of therapy, the potential benefits and risks of corticosteroid treatment should be carefully discussed with each individual.

  • To assess benefits of corticosteroid therapy, the following parameters are useful: timed muscle function tests, pulmonary function tests, and age at loss of independent ambulation. To assess risks of corticosteroid therapy, maintain awareness of the potential corticosteroid therapy side effects (e.g., weight gain, cushingoid appearance, short stature, decrease in linear growth, acne, excessive hair growth, gastrointestinal symptoms, behavioral changes).

  • There is an increased frequency of vertebral and long bone fractures with prolonged corticosteroid use [King et al 2007].

  • The optimal maintenance dose of prednisone (0.75 mg/kg/day) should be continued if side effects are not severe. Significant but less robust improvement can be seen with gradual tapering of prednisone to as low as 0.3 mg/kg/day.

  • If excessive weight gain occurs (>20% over estimated normal weight for height over a 12-month period), the prednisone dose should be decreased to 0.5 mg/kg/day. If excessive weight gain continues, the dose should be further decreased to 0.3 mg/kg/day after three to four months.

  • Deflazacort (0.9 mg/kg/day) can also be used to treat DMD. Side effects of asymptomatic cataracts and weight gain should be monitored.

Information about the efficacy of prednisone in treating individuals with BMD is limited.

Deflazacort. Deflazacort, a synthetic derivative of prednisolone used in Europe but not currently available in the US, is thought to have fewer side effects than prednisone, particularly with regard to weight gain [Angelini 2007]. A larger study comparing deflazacort to prednisone, carried out in Europe, showed that the two medications were similarly or equally effective in slowing the decline of muscle strength in DMD. Another European multicenter, double-blind, randomized trial of deflazacort versus prednisone in DMD showed equal efficacy in improving motor function and functional performance [Bonifati et al 2000]. A more recent study of deflazacort treatment showed efficacy in preserving pulmonary function as well as gross motor function [Biggar et al 2006].

In a comparison of two different protocols of deflazacort treatment in DMD, a 0.9-mg/kg/day dose was more effective than a dose of 0.6 mg/kg/day for the first 20 days of the month and no deflazacort for the remainder of the month [Biggar et al 2004]; 30% of children on the highest dose developed asymptomatic cataracts that required no treatment. A systematic review and meta-analysis of 15 studies showed that deflazacort improves strength and motor function more than placebo; whether it has a benefit over prednisone on similar outcomes remains unclear [Campbell & Jacob 2003].

Scoliosis treatment as needed is appropriate.

Prevention of Secondary Complications

The following measures are appropriate:

  • Evaluation by pulmonary and cardiac specialists before surgeries [Finder et al 2004]

  • Administration of pneumococcal vaccine and influenza vaccination annually [Finder et al 2004]

  • Exposure to sunshine and a balanced diet rich in vitamin D and calcium to improve bone density and reduce the risk of fractures

    Note: Vitamin D supplementation should be initiated if the vitamin D serum concentration is <20 ng/mL [Bachrach 2005, Biggar et al 2005, Quinlivan et al 2005].

  • Physical therapy to promote mobility and prevent contractures

  • Weight control to avoid obesity

    Note: Routine evaluation by a nutritionist is recommended.

Surveillance

Cardiac. The American Academy of Pediatrics (AAP) recommendations for optimal cardiac care in persons with DMD or BMD [American Academy of Pediatrics Section on Cardiology and Cardiac Surgery 2005] include the following:

  • For DMD, complete cardiac evaluation at least every two years, beginning in early childhood

  • For DMD, at approximately age ten years, or at the onset of cardiac signs and symptoms, annual complete cardiac evaluation

    Note: Individuals with DMD demonstrating cardiac signs and symptoms are relatively late in their course.

  • For BMD, complete cardiac evaluations beginning at approximately age ten years or at the onset of signs and symptoms. Evaluations should continue at least every two years.

The AAP recommendations for optimal cardiac care for female carriers of DMD or BMD [American Academy of Pediatrics Section on Cardiology and Cardiac Surgery 2005] include the following:

  • Education about the risk of developing cardiomyopathy and about the signs and symptoms of heart failure

  • Complete cardiac evaluation by a cardiac specialist with experience in the treatment of heart failure and/or neuromuscular disorders, with the initial evaluation to take place in late adolescence or early adulthood, or earlier at the appearance of cardiac signs and symptoms

  • Starting at age 25 to 30 years, screening with a complete cardiac evaluation at least every five years

  • Treatment of cardiac disease similar to that for boys with DMD or BMD

Pulmonary

  • Baseline pulmonary function testing before confinement to a wheelchair (usually age ~9-10 years)

  • Evaluation by a pediatric pulmonologist twice yearly after any one of the following: confinement to a wheelchair, reduction in vital capacity below 80% predicted, and/or age 12 years [Finder et al 2004]

Orthopedic. Monitoring for orthopedic complications, especially scoliosis in those with DMD and BMD

Agents/Circumstances to Avoid

Individuals with DMD/BMD should avoid botulinum toxin injections.

Testing of Relatives at Risk

Females. Female carriers need to be identified for the purpose of cardiac surveillance (see Surveillance).

See Genetic Counseling for issues related to testing of at-risk relatives for genetic counseling purposes.

Therapies Under Investigation

Aminoglycosides. Up to 15% of individuals with DMD exhibit the gene mutation known as a premature stop codon. Suppression of stop codons has been demonstrated with aminoglycoside treatment of cultured cells; the treatment creates misreading of RNA and thereby allows alternative amino acids to be inserted at the site of the mutated stop codon. In the mdx mouse, in vivo gentamicin therapy resulted in dystrophin expression at 10%-20% of that detected in normal muscle [Barton-Davis et al 1999], a level that provided some degree of functional protection against contraction-induced damage.

Aminoglycoside therapy has been suggested as an alternative to gene therapy but could be aimed only at individuals with premature stop codons. In a preliminary study in which gentamicin (7.5 mg/kg/day) was administered to four individuals for two weeks, full-length dystrophin did not appear in the muscles of the treated individuals [Wagner et al 2001]. Some authors, unable to reproduce the results previously published for the mouse model of DMD, have called for more preclinical investigation of this potential therapy [Dunant et al 2003]. In an in vitro study [Kimura et al 2005], dystrophin expression was detected in myotubes of males with DMD using gentamicin; however, the treatment was more effective in persons with the nonsense mutation TGA than TAA or TAG.

PTC124 is a new, orally administered non-antibiotic drug that appears to promote ribosomal read-through of nonsense (stop) mutations. Preclinical efficacy studies in mdx mice have yielded encouraging results [Barton et al 2005, Welch et al 2007]. A Phase I multiple-dose safety trial is ongoing [Hirawat et al 2005].

Morpholino antisense oligonucleotides mediate exon skipping [Aartsma-Rus et al 2006a] and have improved the mdx mouse model of DMD [Wilton & Fletcher 2005, Alter et al 2006].

Oxandrolone, an anabolic (androgenic) steroid with a powerful anabolic effect on skeletal muscle myosin synthesis [Balagopal et al 2006], was shown in a pilot study to have effects similar to prednisone, with fewer side effects [Fenichel et al 1997]. A randomized, prospective, controlled trial showed that oxandrolone did not produce a significant change in the average manual muscle strength score of males with DMD, as compared with placebo; however, the mean change in quantitative muscle strength was significant [Fenichel et al 2001]. The investigators conducting this study felt that oxandrolone may be useful before initiating therapy with corticosteroids because it is safe in the short term, accelerates linear growth, and may be beneficial in slowing the progression of weakness. However, the long-term effects of oxandrolone in the treatment of DMD have not been studied.

Gene therapy. Experimental gene therapies are currently under investigation [Gregorevic & Chamberlain 2003, Tidball & Spencer 2003, van Deutekom & van Ommen 2003, Nowak & Davies 2004].

Gregorevic et al [2004] reported systemic administration of rAAV6 vectors resulting in successful delivery of DMD to affected muscles of dystrophin-deficient mdx mice (a mouse model for DMD).

Stem cell therapy is under investigation but remains experimental [Gussoni et al 1997, Gussoni et al 1999, Gussoni et al 2002, Skuk et al 2004].

Search ClinicalTrials.gov for access to information on clinical studies for a wide range of diseases and conditions.

Other

Immunosuppression with azathioprine is not beneficial.

Myoblast transfer has been inefficient.

Creatine monohydrate has been studied as potential treatment in muscular dystrophies and neuromuscular disorders [Tarnopolsky & Martin 1999, Walter et al 2000, Louis et al 2003]. In a recent randomized, controlled, cross-over trial, 30 boys with DMD were given creatine (~0.1 g/kg/day) for four months and placebo for four months [Tarnopolsky et al 2004]. Treatment with creatine resulted in improved grip strength of the dominant hand and increased fat-free mass when compared to placebo; however, no functional improvement was noted. Given the limited data and modest benefit, treatment with creatine monohydrate cannot be recommended for treatment of DMD.

Cyclosporin was reported to improve clinical function in children with DMD who received the medication for eight weeks. Nevertheless, because of the rare reports of cyclosporin-induced myopathy in individuals receiving the medication for other reasons, the use of cyclosporin in treating DMD remains controversial.

Histone deacetylase inhibitors have improved the mdx mouse by inducing the expression of the myostatin inhibitor follistatin [Minetti et al 2006].

Genetics clinics are a source of information for individuals and families regarding the natural history, treatment, mode of inheritance, and genetic risks to other family members as well as information about available consumer-oriented resources. See the GeneTests Clinic Directory.

Support groups have been established for individuals and families to provide information, support, and contact with other affected individuals. The Resources section may include disease-specific and/or umbrella support organizations.

Genetic Counseling

Genetic counseling is the process of providing individuals and families with information on the nature, inheritance, and implications of genetic disorders to help them make informed medical and personal decisions. The following section deals with genetic risk assessment and the use of family history and genetic testing to clarify genetic status for family members. This section is not meant to address all personal, cultural, or ethical issues that individuals may face or to substitute for consultation with a genetics professional. To find a genetics or prenatal diagnosis clinic, see the GeneTests Clinic Directory.

Mode of Inheritance

The dystrophinopathies are inherited in an X-linked manner.

Risk to Family Members

Parents of a proband

Sibs of a proband

Offspring of a proband

  • Males with DMD usually die before reproductive age or are too debilitated to reproduce.

  • Males with BMD and DMD-associated DCM may reproduce. All the daughters are carriers. None of the sons will inherit their father's DMD mutation.

Other family members of the proband. The proband's maternal aunts and their offspring may be at risk of being carriers or being affected (depending on their gender, family relationship, and the carrier status of the proband's mother).

Carrier Detection

Carrier testing is clinically available for at-risk females. See Molecular genetic testing, Carrier testing.

Related Genetic Counseling Issues

See Testing of Relatives at Risk for information on testing at-risk relatives for the purpose of early diagnosis and treatment.

Females who are identified as carriers of a DMD disease-causing mutation need to be informed of their risk for DCM, as well as the recommended surveillance.

BMD and DMD-associated DCM are sometimes observed in the same family [Palmucci et al 2000]. Thus, the entire spectrum of possible muscle disease should be considered when obtaining a family history and providing genetic counseling.

Family planning

  • The optimal time for determination of genetic risk, clarification of carrier status, and discussion of availability of prenatal testing is before pregnancy.

  • It is appropriate to offer genetic counseling (including discussion of potential risks to offspring and reproductive options) to young adults who are affected or at risk of being carriers.

DNA banking. DNA banking is the storage of DNA (typically extracted from leukocytes) for possible future use. Because it is likely that testing methodology and our understanding of genes, mutations, and diseases will improve in the future, consideration should be given to banking DNA of affected individuals. DNA banking is particularly relevant when the sensitivity of currently available testing is less than 100%. See graphic element for a list of laboratories offering DNA banking.

Prenatal Testing

Prenatal testing is possible for pregnancies of women who are carriers if the DMD mutation has been identified in a family member or if linkage has been established. The usual procedure is to determine fetal sex by karyotype or specialized studies to identify the sex chromosomes from cells obtained by chorionic villus sampling (CVS) at approximately ten to 12 weeks' gestation or by amniocentesis usually performed at approximately 15-18 weeks' gestation. If the karyotype is 46,XY, DNA extracted from fetal cells can be analyzed for the known disease-causing mutation or using the linkage previously established.

Note: Gestational age is expressed as menstrual weeks calculated either from the first day of the last normal menstrual period or by ultrasound measurements.

Preimplantation genetic diagnosis (PGD). Preimplantation genetic diagnosis may be available for families in which the disease-causing mutation has been identified. For laboratories offering PGD, see graphic element.

Molecular Genetics

Information in the Molecular Genetics tables is current as of initial posting or most recent update. —ED.

Table A. Molecular Genetics of Dystrophinopathies

Gene SymbolChromosomal LocusProtein Name
DMDXp21.2Dystrophin

Data are compiled from the following standard references: gene symbol from HUGO; chromosomal locus, locus name, critical region, complementation group from OMIM; protein name from Swiss-Prot.

Table B. OMIM Entries for Dystrophinopathies

300376 MUSCULAR DYSTROPHY, BECKER TYPE; BMD
300377 DYSTROPHIN; DMD
302045 CARDIOMYOPATHY, DILATED, 3B; CMD3B
310200 MUSCULAR DYSTROPHY, DUCHENNE TYPE; DMD

Table C. Genomic Databases for Dystrophinopathies

Gene SymbolLocus SpecificEntrez GeneHGMD
DMDDMD1756 (MIM No. 300377)DMD

For a description of the genomic databases listed, click here.

Note: HGMD requires registration.

Normal allelic variants. The DMD gene spans 2.4 Mb of DNA and comprises 79 exons. It has at least four promoters. It is the largest known human gene. Innumerable intragenic variants have been described, many of which are useful as markers for genetic linkage analysis.

Pathologic allelic variants. Disease-causing alleles are highly variable, including deletion of the entire gene, deletion or duplication of one or more exons, and small deletions, insertions, or single-base changes. In both DMD and BMD, partial deletions and duplications cluster in two recombination hot spots, one proximal at the 5' end of the gene, comprising exons 2-20 (30%), and one more distal, comprising exons 44-53 (70%) [Den Dunnen et al 1989]. Duplications cluster near the 5' end of the gene, with duplication of exon 2 being the single most common duplication identified [White et al 2006]. More than 4,700 mutations have been identified [Aartsma-Rus et al 2006b].

Normal gene product. Dystrophin is a membrane-associated protein present in muscle cells and some neurons. The N-terminal domain binds to actin. A large rod domain includes 24 homologous repeats forming an α-helical structure, a cysteine-rich calcium-binding region near the C terminus, and a C-terminal domain that binds with other membrane proteins. Dystrophin is therefore part of a protein complex that links the cytoskeleton with membrane proteins that in turn bind with proteins in the extracellular matrix.

Abnormal gene product. Mutations that lead to lack of dystrophin expression tend to cause DMD, whereas those that lead to abnormal quality or quantity of dystrophin lead to BMD. In DMD-associated DCM, dystrophin expression is abnormal in the myocardium and may be normal or mildly abnormal in skeletal muscle [Ferlini et al 1999].

Resources

GeneReviews provides information about selected national organizations and resources for the benefit of the reader. GeneReviews is not responsible for information provided by other organizations. Information that appears in the Resources section of a GeneReview is current as of initial posting or most recent update of the GeneReview. Search GeneTests for this disorder and select graphic element for the most up-to-date Resources information.—ED.

National Library of Medicine Genetics Home Reference
Muscular dystrophy, Duchenne and Becker types

NCBI Genes and Disease
Duchenne muscular dystrophy

Parent Project Muscular Dystrophy
1012 North University Boulevard
Middletown OH 45042
Phone: 800-714-5437; 513-424-0696
Fax: 513-425-9907
Email: pat@parentprojectmd.org
www.parentprojectmd.org

European Neuromuscular Centre (ENMC)
Lt. Gen. van Heutszlaan 6
3743 JN Baarn
Netherlands
Phone: 035 54 80 481
Fax: 035 54 80 499
Email: info@enmc.org
www.enmc.org

Medline Plus
Muscular Dystrophy

Muscular Dystrophy Association (MDA)
3300 East Sunrise Drive
Tucson AZ 85718-3208
Phone: 800-FIGHT-MD (800-344-4863); 520-529-2000
Fax: 520-529-5300
Email: mda@mdausa.org
www.mdausa.org

Muscular Dystrophy Association-Canada
2345 Yonge Street Suite 900
Toronto M4P 2E5
Canada
Phone: 866-MUSCLE-8 (866-687-2538); 416-488-0030
Fax: 416-488-7523
Email: info@muscle.ca
www.muscle.ca

Muscular Dystrophy Campaign
7-11 Prescott Place
SW4 6BS
United Kingdom
Phone: (+44) 0 020 7720 8055
Fax: (+44) 0 020 7498 0670
Email: info@muscular-dystrophy.org
www.muscular-dystrophy.org

DuchenneConnect
DuchenneConnect has been created to serve as a central hub linking the resources and needs of the Duchenne/Becker MD community. One goal of their Registry is to develop new and improved treatments and learn more about the impact of Duchenne/Becker muscular dystrophy on individuals and families.
Phone: 404-778-0553
Fax: 404-778-8559
Email: coordinator@duchenneconnect.org
www.duchenneconnect.org

Teaching Case-Genetic Tools
Cases designed for teaching genetics in the primary care setting.
Case 23. A Young Woman with a Family History of Duchenne Muscular Dystrophy

References

Medical Genetic Searches: A specialized PubMed search designed for clinicians that is located on the PubMed Clinical Queries page. graphic element

Literature Cited

Aartsma-Rus A, Janson AA, Heemskerk JA, De Winter CL, Van Ommen GJ, Van Deutekom JC. Therapeutic modulation of DMD splicing by blocking exonic splicing enhancer sites with antisense oligonucleotides. Ann N Y Acad Sci. 2006a; 1082: 746. [PubMed]
Aartsma-Rus A, Van Deutekom JC, Fokkema IF, Van Ommen GJ, Den Dunnen JT. Entries in the Leiden Duchenne muscular dystrophy mutation database: an overview of mutation types and paradoxical cases that confirm the reading-frame rule. Muscle Nerve. 2006b; 34: 13544. [PubMed]
Abbs S, Roberts RG, Mathew CG, Bentley DR, Bobrow M. Accurate assessment of intragenic recombination frequency within the Duchenne muscular dystrophy gene. Genomics. 1990; 7: 6026. [PubMed]
Alter J, Lou F, Rabinowitz A, Yin H, Rosenfeld J, Wilton SD, Partridge TA, Lu QL. Systemic delivery of morpholino oligonucleotide restores dystrophin expression bodywide and improves dystrophic pathology. Nat Med. 2006; 12: 1757. [PubMed]
American Academy of Pediatrics Section on Cardiology and Cardiac Surgery; Clinical Report: cardiovascular health supervision for individuals affected by Duchenne or Becker muscular dystrophy. Pediatrics. 2005; 116: 156973. [PubMed]
Angelini C. The role of corticosteroids in muscular dystrophy: a critical appraisal. Muscle Nerve. 2007; 36: 42435. [PubMed]
Bachrach LK. Taking steps towards reducing osteoporosis in Duchenne muscular dystrophy. Neuromuscul Disord. 2005; 15: 867. [PubMed]
Bakker E, Van Broeckhoven C, Bonten EJ, van de Vooren MJ, Veenema H, Van Hul W, Van Ommen GJ, Vandenberghe A, Pearson PL. Germline mosaicism and Duchenne muscular dystrophy mutations. Nature. 1987; 329: 5546. [PubMed]
Balagopal P, Olney R, Darmaun D, Mougey E, Dokler M, Sieck G, Hammond D. Oxandrolone enhances skeletal muscle myosin synthesis and alters global gene expression profile in Duchenne muscular dystrophy. Am J Physiol Endocrinol Metab. 2006; 290: E5309. [PubMed]
Barton E, Zadel M, Welch E, Rotta C, Paushkin S, Patel M, Zhuo J, Tomizawa Y, Wectall M, Northcutt V, Bahiak J, Miller L, Sweeney L. PTC124 nonsense mutation suppression therapy of Duchenne muscular dystrophy (DMD). Neurology. 2005; 54 Suppl 1: A176.
Barton-Davis ER, Cordier L, Shoturma DI, Leland SE, Sweeney HL. Aminoglycoside antibiotics restore dystrophin function to skeletal muscles of mdx mice. J Clin Invest. 1999; 104: 37581. [PubMed]
Beenakker EA, Fock JM, Van Tol MJ, Maurits NM, Koopman HM, Brouwer OF, Van der Hoeven JH. Intermittent prednisone therapy in Duchenne muscular dystrophy: a randomized controlled trial. Arch Neurol. 2005; 62: 12832. [PubMed]
Beggs AH. Dystrophinopathy, the expanding phenotype. Dystrophin abnormalities in X-linked dilated cardiomyopathy. Circulation. 1997; 95: 23447. [PubMed]
Bennett RR, den Dunnen J, O'Brien KF, Darras BT, Kunkel LM. Detection of mutations in the dystrophin gene via automated DHPLC screening and direct sequencing. BMC Genet. 2001; 2: 17. [PubMed]
Biggar WD, Bachrach LK, Henderson RC, Kalkwarf H, Plotkin H, Wong BL. Bone health in Duchenne muscular dystrophy: a workshop report from the meeting in Cincinnati, Ohio, July 8, 2004. Neuromuscul Disord. 2005; 15: 805. [PubMed]
Biggar WD, Harris VA, Eliasoph L, Alman B. Long-term benefits of deflazacort treatment for boys with Duchenne muscular dystrophy in their second decade. Neuromuscul Disord. 2006; 16: 24955. [PubMed]
Biggar WD, Politano L, Harris VA, Passamano L, Vajsar J, Alman B, Palladino A, Comi LI, Nigro G. Deflazacort in Duchenne muscular dystrophy: a comparison of two different protocols. Neuromuscul Disord. 2004; 14: 47682. [PubMed]
Bonifati MD, Ruzza G, Bonometto P, Berardinelli A, Gorni K, Orcesi S, Lanzi G, Angelini C. A multicenter, double-blind, randomized trial of deflazacort versus prednisone in Duchenne muscular dystrophy. Muscle Nerve. 2000; 23: 13447. [PubMed]
Bushby K, Muntoni F, Urtizberea A, Hughes R, Griggs R. Report on the 124th ENMC International Workshop. Treatment of Duchenne muscular dystrophy; defining the gold standards of management in the use of corticosteroids. 2-4 April 2004, Naarden, The Netherlands. Neuromuscul Disord. 2004; 14: 52634. [PubMed]
Bushby KM. The limb-girdle muscular dystrophies—multiple genes, multiple mechanisms. Hum Mol Genet. 1999; 8: 187582. [PubMed]
Campbell C, Jacob P. Deflazacort for the treatment of Duchenne Dystrophy: a systematic review. BMC Neurol. 2003; 3: 7. [PubMed]
Chelly J, Gilgenkrantz H, Lambert M, Hamard G, Chafey P, Recan D, Katz P, de la Chapelle A, Koenig M, Ginjaar IB. et al. Effect of dystrophin gene deletions on mRNA levels and processing in Duchenne and Becker muscular dystrophies. Cell. 1990; 63: 123948. [PubMed]
Colan SD. Evolving therapeutic strategies for dystrophinopathies: potential for conflict between cardiac and skeletal needs. Circulation. 2005; 112: 27568. [PubMed]
Connolly AM, Schierbecker J, Renna R, Florence J. High dose weekly oral prednisone improves strength in boys with Duchenne muscular dystrophy. Neuromuscul Disord. 2002; 12: 91725. [PubMed]
Cox GF, Kunkel LM. Dystrophies and heart disease. Curr Opin Cardiol. 1997; 12: 32943. [PubMed]
D'Adamo P, Fassone L, Gedeon A, Janssen EA, Bione S, Bolhuis PA, Barth PG, Wilson M, Haan E, Orstavik KH, Patton MA, Green AJ, Zammarchi E, Donati MA, Toniolo D. The X-linked gene G4.5 is responsible for different infantile dilated cardiomyopathies. Am J Hum Genet. 1997; 61: 8627. [PubMed]
Darras BT, Francke U. A partial deletion of the muscular dystrophy gene transmitted twice by an unaffected male. Nature. 1987; 329: 5568. [PubMed]
Darras BT, Francke U. Myopathy in complex glycerol kinase deficiency patients is due to 3' deletions of the dystrophin gene. Am J Hum Genet. 1988; 43: 12630. [PubMed]
Darras BT, Koenig M, Kunkel LM, Francke U. Direct method for prenatal diagnosis and carrier detection in Duchenne/Becker muscular dystrophy using the entire dystrophin cDNA. Am J Med Genet. 1988; 29: 71326. [PubMed]
Deburgrave N, Daoud F, Llense S, Barbot JC, Recan D, Peccate C, Burghes AH, Beroud C, Garcia L, Kaplan JC, Chelly J, Leturcq F. Pro. Hum Mutat. 2007; 28: 18395. [PubMed]
Den Dunnen JT, Grootscholten PM, Bakker E, Blonden LA, Ginjaar HB, Wapenaar MC, van Paassen HM, van Broeckhoven C, Pearson PL, van Ommen GJ. Topography of the Duchenne muscular dystrophy (DMD) gene: FIGE and cDNA analysis of 194 cases reveals 115 deletions and 13 duplications. Am J Hum Genet. 1989; 45: 83547. [PubMed]
Dolinsky LC, de Moura-Neto RS, Falcao-Conceicao DN. DGGE analysis as a tool to identify point mutations, de novo mutations and carriers of the dystrophin gene. Neuromuscul Disord. 2002; 12: 8458. [PubMed]
Duboc D, Meune C, Lerebours G, Devaux JY, Vaksmann G, Becane HM. Effect of perindopril on the onset and progression of left ventricular dysfunction in Duchenne muscular dystrophy. J Am Coll Cardiol. 2005; 45: 8557. [PubMed]
Dubowitz V. 47th ENMC international workshop: Treatment of muscular dystrophy. Neuromusc Disord. 1997; 7: 2617. [PubMed]
Dunant P, Walter MC, Karpati G, Lochmuller H. Gentamicin fails to increase dystrophin expression in dystrophin-deficient muscle. Muscle Nerve. 2003; 27: 6247. [PubMed]
Felisari G, Martinelli Boneschi F, Bardoni A, Sironi M, Comi GP, Robotti M, Turconi AC, Lai M, Corrao G, Bresolin N. Loss of Dp140 dystrophin isoform and intellectual impairment in Duchenne dystrophy. Neurology. 2000; 55: 55964. [PubMed]
Fenichel G, Pestronk A, Florence J, Robison V, Hemelt V. A beneficial effect of oxandrolone in the treatment of Duchenne muscular dystrophy: a pilot study. Neurology. 1997; 48: 12256. [PubMed]
Fenichel GM, Florence JM, Pestronk A, Mendell JR, Moxley RT, Griggs RC, Brooke MH, Miller JP, Robison J, King W. et al. Long-term benefit from prednisone therapy in Duchenne muscular dystrophy. Neurology. 1991; 41: 18747. [PubMed]
Fenichel GM, Griggs RC, Kissel J, Kramer TI, Mendell JR, Moxley RT, Pestronk A, Sheng K, Florence J, King WM, Pandya S, Robison VD, Wang H. A randomized efficacy and safety trial of oxandrolone in the treatment of Duchenne dystrophy. Neurology. 2001; 56: 10759. [PubMed]
Ferlini A, Sewry C, Melis MA, Mateddu A, Muntoni F. X-linked dilated cardiomyopathy and the dystrophin gene. Neuromuscul Disord. 1999; 9: 33946. [PubMed]
Ferreiro V, Szijan I, Giliberto F. Detection of germline mosaicism in two Duchenne muscular dystrophy families using polymorphic dinucleotide (CA)n repeat loci within the dystrophin gene. Mol Diagn. 2004; 8: 11521. [PubMed]
Finder JD, Birnkrant D, Carl J, Farber HJ, Gozal D, Iannaccone ST, Kovesi T, Kravitz RM, Panitch H, Schramm C, Schroth M, Sharma G, Sievers L, Silvestri JM, Sterni L. Respiratory care of the patient with Duchenne muscular dystrophy: ATS consensus statement. Am J Respir Crit Care Med. 2004; 170: 45665. [PubMed]
Finsterer J, Stollberger C. The heart in human dystrophinopathies. Cardiology. 2003; 99: 119. [PubMed]
Flanigan KM, von Niederhausern A, Dunn DM, Alder J, Mendell JR, Weiss RB. Rapid direct sequence analysis of the dystrophin gene. Am J Hum Genet. 2003; 72: 9319. [PubMed]
Francke U, Ochs HD, de Martinville B, Giacalone J, Lindgren V, Disteche C, Pagon RA, Hofker MH, van Ommen GJ, Pearson PL. et al. Minor Xp21 chromosome deletion in a male associated with expression of Duchenne muscular dystrophy, chronic granulomatous disease, retinitis pigmentosa, and McLeod syndrome. Am J Hum Genet. 1985; 37: 25067. [PubMed]
Galvagni F, Saad FA, Danieli GA, Miorin M, Vitiello L, Mostacciuolo ML, Angelini C. A study on duplications of the dystrophin gene: evidence of a geographical difference in the distribution of breakpoints by intron. Hum Genet. 1994; 94: 837. [PubMed]
Gatta V, Scarciolla O, Gaspari AR, Palka C, De Angelis MV, Di Muzio A, Guanciali-Franchi P, Calabrese G, Uncini A, Stuppia L. Identification of deletions and duplications of the DMD gene in affected males and carrier females by multiple ligation probe amplification (MLPA). Hum Genet. 2005; 117: 928. [PubMed]
Gavillet B, Rougier JS, Domenighetti AA, Behar R, Boixel C, Ruchat P, Lehr HA, Pedrazzini T, Abriel H. Cardiac sodium channel Nav1.5 is regulated by a multiprotein complex composed of syntrophins and dystrophin. Circ Res. 2006; 99: 40714. [PubMed]
Gregorevic P, Blankinship MJ, Allen JM, Crawford RW, Meuse L, Miller DG, Russell DW, Chamberlain JS. Systemic delivery of genes to striated muscles using adeno-associated viral vectors. Nat Med. 2004; 10: 82834. [PubMed]
Gregorevic P, Chamberlain JS. Gene therapy for muscular dystr. Expert Opin Biol Ther. 2003; 3: 80314. [PubMed]
Griggs RC, Moxley RT, Mendell JR, Fenichel GM, Brooke MH, Pestronk A, Miller JP, Cwik VA, Pandya S, Robison J. et al. Duchenne dystrophy: randomized, controlled trial of prednisone (18 months) and azathioprine (12 months). Neurology. 1993; 43: 5207. [PubMed]
Gussoni E, Bennett RR, Muskiewicz KR, Meyerrose T, Nolta JA, Gilgoff I, Stein J, Chan YM, Lidov HG, Bonnemann CG, Von Moers A, Morris GE, Den Dunnen JT, Chamberlain JS, Kunkel LM, Weinberg K. Long-term persistence of donor nuclei in a Duchenne muscular dystrophy patient receiving bone marrow transplantation. J Clin Invest. 2002; 110: 80714. [PubMed]
Gussoni E, Blau HM, Kunkel LM. The fate of individual myoblasts after transplantation into muscles of DMD patients. Nat Med. 1997; 3: 9707. [PubMed]
Gussoni E, Soneoka Y, Strickland CD, Buzney EA, Khan MK, Flint AF, Kunkel LM, Mulligan RC. Dystrophin expression in the mdx mouse restored by stem cell transplantation. Nature. 1999; 401: 3904. [PubMed]
Hinton VJ, De Vivo DC, Nereo NE, Goldstein E, Stern Y. Selective deficits in verbal working memory associated with a known genetic etiology: the neuropsychological profile of duchenne muscular dystrophy. J Int Neuropsychol Soc. 2001; 7: 4554. [PubMed]
Hirawat S, Northcutt VJ, Welch EM, Elfring GL, Hwang S, Almstead NG, Ju W, Miller LL. Phase 1 multiple-dose safety and PK study of PTC124 for nonsense mutation suppression therapy of Duchenne muscular dystrophy (DMD). Neurology. 2005; 54 Suppl 1: A176.
Hoffman EP, Fischbeck KH, Brown RH, Johnson M, Medori R, Loike JD, Harris JB, Waterston R, Brooke M, Specht L. et al. Characterization of dystrophin in muscle-biopsy specimens from patients with Duchenne's or Becker's muscular dystrophy. N Engl J Med. 1988; 318: 13638. [PubMed]
Hoffman EP, Garcia CA, Chamberlain JS, Angelini C, Lupski JR, Fenwick R. Is the carboxyl-terminus of dystrophin required for membrane association? A novel, severe case of Duchenne muscular dystrophy. Ann Neurol. 1991; 30: 60510. [PubMed]
Hofstra RM, Mulder IM, Vossen R, de Koning-Gans PA, Kraak M, Ginjaar IB, van der Hout AH, Bakker E, Buys CH, van Ommen GJ, van Essen AJ, den Dunnen JT. DGGE-based whole-gene mutation scanning of the dystrophin gene in Duchenne and Becker muscular dystrophy patients. Hum Mutat. 2004; 23: 5766. [PubMed]
Hoogerwaard EM, Bakker E, Ippel PF, Oosterwijk JC, Majoor-Krakauer DF, Leschot NJ, Van Essen AJ, Brunner HG, van der Wouw PA, Wilde AA, de Visser M. Signs and symptoms of Duchenne muscular dystrophy and Becker muscular dystrophy among carriers in The Netherlands: a cohort study. Lancet. 1999a; 353: 21169. [PubMed]
Hoogerwaard EM, van der Wouw PA, Wilde AA, Bakker E, Ippel PF, Oosterwijk JC, Majoor-Krakauer DF, van Essen AJ, Leschot NJ, de Visser M. Cardiac involvement in carriers of Duchenne and Becker muscular dystrophy. Neuromuscul Disord. 1999b; 9: 34751. [PubMed]
Jefferies JL, Eidem BW, Belmont JW, Craigen WJ, Ware SM, Fernbach SD, Neish SR, Smith EO, Towbin JA. Genetic predictors and remodeling of dilated cardiomyopathy in muscular dystrophy. Circulation. 2005; 112: 2799804. [PubMed]
Joncourt F, Neuhaus B, Jostarndt-Foegen K, Kleinle S, Steiner B, Gallati S. Rapid identification of female carriers of DMD/BMD by quantitative real-time PCR. Hum Mutat. 2004; 23: 38591. [PubMed]
Keeling RM, Golumbek PT, Streif EM, Connolly AM. Weekly oral prednisolone improves survival and strength in male mdx mice. Muscle Nerve. 2007; 35: 438. [PubMed]
Kim UK, Chae JJ, Lee SH, Lee CC, Namkoong Y. Molecular diagnosis of Duchenne/Becker muscular dystrophy by polymerase chain reaction and microsatellite analysis. Mol Cells. 2002; 13: 3858. [PubMed]
Kimura S, Ito K, Miyagi T, Hiranuma T, Yoshioka K, Ozasa S, Matsukura M, Ikezawa M, Matsuo M, Takeshima Y, Miike T. A novel approach to identify Duchenne muscular dystrophy patients for aminoglycoside antibiotics therapy. Brain Dev. 2005; 27: 4005. [PubMed]
King WM, Ruttencutter R, Nagaraja HN, Matkovic V, Landoll J, Hoyle C, Mendell JR, Kissel JT. Orthopedic outcomes of long-term daily corticosteroid treatment in Duchenne muscular dystrophy. Neurology. 2007; 68: 160713. [PubMed]
Koenig M, Beggs AH, Moyer M, Scherpf S, Heindrich K, Bettecken T, Meng G, Muller CR, Lindlof M, Kaariainen H. et al. The molecular basis for Duchenne versus Becker muscular dystrophy: correlation of severity with type of deletion. Am J Hum Genet. 1989; 45: 498506. [PubMed]
Louis M, Lebacq J, Poortmans JR, Belpaire-Dethiou MC, Devogelaer JP, Van Hecke P, Goubel F, Francaux M. Beneficial effects of creatine supplementation in dystrophic patients. Muscle Nerve. 2003; 27: 60410. [PubMed]
Manzur AY, Kuntzer T, Pike M, Swan A. Glucocorticoid corticosteroids for Duchenne muscular dystrophy. Cochrane Database Syst Rev. 2004; (2): CD003725. [PubMed]
Melacini P, Fanin M, Danieli GA, Villanova C, Martinello F, Miorin M, Freda MP, Miorelli M, Mostacciuolo ML, Fasoli G, Angelini C, Dalla Volta S. Myocardial involvement is very frequent among patients affected with subclinical Becker's muscular dystrophy. Circulation. 1996; 94: 316875. [PubMed]
Mendell JR, Buzin CH, Feng J, Yan J, Serrano C, Sangani DS, Wall C, Prior TW, Sommer SS. Diagnosis of Duchenne dystrophy by enhanced detection of small mutations. Neurology. 2001; 57: 64550. [PubMed]
Mendell JR, Moxley RT, Griggs RC, Brooke MH, Fenichel GM, Miller JP, King W, Signore L, Pandya S, Florence J. et al. Randomized, double-blind six-month trial of prednisone in Duchenne's muscular dystrophy. N Engl J Med. 1989; 320: 15927. [PubMed]
Merlini L, Cicognani A, Malaspina E, Gennari M, Gnudi S, Talim B, Franzoni E. Early prednisone treatment in Duchenne muscular dystrophy. Muscle Nerve. 2003; 27: 2227. [PubMed]
Mestroni L, Rocco C, Gregori D, Sinagra G, Di Lenarda A, Miocic S, Vatta M, Pinamonti B, Muntoni F, Caforio AL, McKenna WJ, Falaschi A, Giacca M. Familial dilated cardiomyopathy: evidence for genetic and phenotypic heterogeneity. Heart Muscle Disease Study Group. J Am Coll Cardiol. 1999; 34: 18190. [PubMed]
Minetti GC, Colussi C, Adami R, Serra C, Mozzetta C, Parente V, Fortuni S, Straino S, Sampaolesi M, Di Padova M, Illi B, Gallinari P, Steinkuhler C, Capogrossi MC, Sartorelli V, Bottinelli R, Gaetano C, Puri PL. Functional and morphological recovery of dystrophic muscles in mice treated with deacetylase inhibitors. Nat Med. 2006; 12: 114750. [PubMed]
Monaco AP, Bertelson CJ, Liechti-Gallati S, Moser H, Kunkel LM. An explanation for the phenotypic differences between patients bearing partial deletions of the DMD locus. Genomics. 1988; 2: 905. [PubMed]
Moxley RT 3rd, Ashwal S, Pandya S, Connolly A, Florence J, Mathews K, Baumbach L, McDonald C, Sussman M, Wade C. Practice parameter: corticosteroid treatment of Duchenne dystrophy: report of the Quality Standards Subcommittee of the American Academy of Neurology and the Practice Committee of the Child Neurology Society. Neurology. 2005; 64: 1320. [PubMed]
Multicenter Study Group; Diagnosis of Duchenne and Becker muscular dystrophies by polymerase chain reaction. JAMA. 1992; 267: 260915. [PubMed]
Nigro G, Comi LI, Politano L, Bain RJ. The incidence and evolution of cardiomyopathy in Duchenne muscular dystrophy. Int J Cardiol. 1990; 26: 2717. [PubMed]
Nolan MA, Jones OD, Pedersen RL, Johnston HM. Cardiac assessment in childhood carriers of Duchenne and Becker muscular dystrophies. Neuromuscul Disord. 2003; 13: 12932. [PubMed]
Nowak KJ, Davies KE. Duchenne muscular dystrophy and dystrophin: pathogenesis and opportunities for treatment. EMBO Rep. 2004; 5: 8726. [PubMed]
Ortiz-Lopez R, Li H, Su J, Goytia V, Towbin JA. Evidence for a dystrophin missense mutation as a cause of X-linked dilated cardiomyopathy. Circulation. 1997; 95: 243440. [PubMed]
Palmucci L, Mongini T, Chiado-Piat L, Doriguzzi C, Fubini A. Dystrophinopathy expressing as either cardiomyopathy or Becker dystrophy in the same family. Neurology. 2000; 54: 52930. [PubMed]
Parker AE, Robb SA, Chambers J, Davidson AC, Evans K, O'Dowd J, Williams AJ, Howard RS. Analysis of an adult Duchenne muscular dystrophy population. QJM. 2005; 98: 72936. [PubMed]
Pegoraro E, Schimke RN, Garcia C, Stern H, Cadaldini M, Angelini C, Barbosa E, Carroll J, Marks WA, Neville HE, Marks H, Appleton S, Toriello H, Wessel HB, Donnelly J, Bernes SM, Taber JW, Weiss L, Hoffman EP. Genetic and biochemical normalization in female carriers of Duchenne muscular dystrophy: evidence for failure of dystrophin production in dystrophin-competent myonuclei. Neurology. 1995; 45: 67790. [PubMed]
Quinlivan R, Roper H, Davie M, Shaw NJ, McDonagh J, Bushby K. Report of a Muscular Dystrophy Campaign funded workshop Birmingham, UK, January 16th 2004. Osteoporosis in Duchenne muscular dystrophy; its prevalence, treatment and prevention. Neuromuscul Disord. 2005; 15: 729. [PubMed]
Saito M, Kawai H, Akaike M, Adachi K, Nishida Y, Saito S. Cardiac dysfunction with Becker muscular dystrophy. Am Heart J. 1996; 132: 6427. [PubMed]
Schwartz M, Hertz JM, Sveen ML, Vissing J. LGMD2I presenting with a characteristic Duchenne or Becker muscular dystrophy phenotype. Neurology. 2005; 64: 16357. [PubMed]
Skuk D, Roy B, Goulet M, Chapdelaine P, Bouchard JP, Roy R, Dugre FJ, Lachance JG, Deschenes L, Helene S, Sylvain M, Tremblay JP. Dystrophin expression in myofibers of Duchenne muscular dystrophy patients following intramuscular injections of normal myogenic cells. Mol Ther. 2004; 9: 47582. [PubMed]
Sumita DR, Vainzof M, Campiotto S, Cerqueira AM, Canovas M, Otto PA, Passos-Bueno MR, Zatz M. Absence of correlation between skewed X inactivation in blood and serum creatine-kinase levels in Duchenne/Becker female carriers. Am J Med Genet. 1998; 80: 35661. [PubMed]
Tarnopolsky M, Martin J. Creatine monohydrate increases strength in patients with neuromuscular disease. Neurology. 1999; 52: 8547. [PubMed]
Tarnopolsky MA, Mahoney DJ, Vajsar J, Rodriguez C, Doherty TJ, Roy BD, Biggar D. Creatine monohydrate enhances strength and body composition in Duchenne muscular dystrophy. Neurology. 2004; 62: 17717. [PubMed]
Tidball JG, Spencer MJ. Skipping to new gene therapies for muscular dystrophy. Nat Med. 2003; 9: 9978. [PubMed]
Towbin JA. A noninvasive means of detecting preclinical cardiomyopathy in Duchenne muscular dystrophy? J Am Coll Cardiol. 2003; 42: 3178. [PubMed]
Towbin JA. The role of cytoskeletal proteins in cardiomyopathies. Curr Opin Cell Biol. 1998; 10: 1319. [PubMed]
Towbin JA, Hejtmancik JF, Brink P, Gelb B, Zhu XM, Chamberlain JS, McCabe ER, Swift M. X-linked dilated cardiomyopathy. Molecular genetic evidence of linkage to the Duchenne muscular dystrophy (dystrophin) gene at the Xp21 locus. Circulation. 1993; 87: 185465. [PubMed]
van Deutekom JC, van Ommen GJ. Advances in Duchenne muscular dystrophy gene therapy. Nat Rev Genet. 2003; 4: 77483. [PubMed]
van Essen AJ, Busch HF, te Meerman GJ, ten Kate LP. Birth and population prevalence of Duchenne muscular dystrophy in The Netherlands. Hum Genet. 1992; 88: 25866. [PubMed]
van Essen AJ, Kneppers AL, van der Hout AH, Scheffer H, Ginjaar IB, ten Kate LP, van Ommen GJ, Buys CH, Bakker E. The clinical and molecular genetic approach to Duchenne and Becker muscular dystrophy: an updated protocol. J Med Genet. 1997; 34: 80512. [PubMed]
van Essen AJ, Mulder IM, van der Vlies P, van der Hout AH, Buys CH, Hofstra RM, den Dunnen JT. Detection of point mutation in dystrophin gene reveals somatic and germline mosaicism in the mother of a patient with Duchenne muscular dystrophy. Am J Med Genet. 2003; 118A: 2968.
Voskova-Goldman A, Peier A, Caskey CT, Richards CS, Shaffer LG. DMD-specific FISH probes are diagnostically useful in the detection of female carriers of DMD gene deletions. Neurology. 1997; 48: 16338. [PubMed]
Wagner KR, Hamed S, Hadley DW, Gropman AL, Burstein AH, Escolar DM, Hoffman EP, Fischbeck KH. Gentamicin treatment of Duchenne and Becker muscular dystrophy due to nonsense mutations. Ann Neurol. 2001; 49: 70611. [PubMed]
Walter MC, Lochmuller H, Reilich P, Klopstock T, Huber R, Hartard M, Hennig M, Pongratz D, Muller-Felber W. Creatine monohydrate in muscular dystrophies: A double-blind, placebo-controlled clinical study. Neurology. 2000; 54: 184850. [PubMed]
Welch EM, Barton ER, Zhuo J, Tomizawa Y, Friesen WJ, Trifillis P, Paushkin S, Patel M, Trotta CR, Hwang S, Wilde RG, Karp G, Takasugi J, Chen G, Jones S, Ren H, Moon YC, Corson D, Turpoff AA, Campbell JA, Conn MM, Khan A, Almstead NG, Hedrick J, Mollin A, Risher N, Weetall M, Yeh S, Branstrom AA, Colacino JM, Babiak J, Ju WD, Hirawat S, Northcutt VJ, Miller LL, Spatrick P, He F, Kawana M, Feng H, Jacobson A, Peltz SW, Sweeney HL. PTC124 targets genetic disorders caused by nonsense mutations. Nature. 2007; 447: 8791. [PubMed]
White SJ, Aartsma-Rus A, Flanigan KM, Weiss RB, Kneppers AL, Lalic T, Janson AA, Ginjaar HB, Breuning MH, den Dunnen JT. Duplications in the DMD gene. Hum Mutat. 2006; 27: 93845. [PubMed]
Wicksell RK, Kihlgren M, Melin L, Eeg-Olofsson O. Specific cognitive deficits are common in children with Duchenne muscular dystrophy. Dev Med Child Neurol. 2004; 46: 1549. [PubMed]
Wilton SD, Fletcher S. Antisense oligonucleotides, exon skipping and the dystrophin gene transcript. Acta Myol. 2005; 24: 2229. [PubMed]
Yazaki M, Yoshida K, Nakamura A, Koyama J, Nanba T, Ohori N, Ikeda S. Clinical characteristics of aged Becker muscular dystrophy patients with onset after 30 years. Eur Neurol. 1999; 42: 1459. [PubMed]
Yen TY, Hwu WL, Chien YH, Wu MH, Lin MT, Tsao LY, Hsieh WS, Lee NC. Acute metabolic decompensation and sudden death in Barth syndrome: report of a family and a literature review. Eur J Pediatr. 2008; 167: 9414. [PubMed]
Yoshida K, Nakamura A, Yazaki M, Ikeda S, Takeda S. Insertional mutation by transposable element, L1, in the DMD gene results in X-linked dilated cardiomyopathy. Hum Mol Genet. 1998; 7: 112932. [PubMed]
Zalaudek I, Bonelli RM, Koltringer P, Reisecker F, Wagner K. Early diagnosis in Duchenne muscular dystrophy. Lancet. 1999; 353: 1975. [PubMed]
Zatz M, Rapaport D, Vainzof M, Passos-Bueno MR, Bortolini ER, Pavanello R de C, Peres CA. Serum creatine-kinase (CK) and pyruvate-kinase (PK) activities in Duchenne (DMD) as compared with Becker (BMD) muscular dystrophy. J Neurol Sci. 1991; 102: 1906. [PubMed]

Published Statements and Policies Regarding Genetic Testing

No specific guidelines regarding genetic testing for this disorder have been developed.

Suggested Reading

Kang PB, Kunkel LM. The muscular dystrophies. In: Scriver CR, Beaudet AL, Sly WS, Valle D, Vogelstein B (eds) The Metabolic and Molecular Bases of Inherited Disease (OMMBID), McGraw-Hill, New York, Chap 216. Available at www.ommbid.com. Accessed 3-6-08.

Chapter Notes

Revision History

  • 21 March 2008 (me) Comprehensive update posted to live Web site

  • 25 August 2005 (me) Comprehensive update posted to live Web site

  • 1 October 2004 (cd) Revision

  • 3 August 2004 (cd) Revision: Management

  • 24 March 2004 (cd) Revision: Diagnosis

  • 23 June 2003 (me) Comprehensive update posted to live Web site

  • 5 September 2000 (me) Review posted to live Web site

  • December 1999 (bk) Original submission

Next
GeneReviews2009
(navigation arrows) Go to previous chapter Go to next chapter Go to top of this page Go to bottom of this page Go to Table of Contents