Muscular Dystrophy

Earn CME/CE in your profession:


Continuing Education Activity

The term "muscular dystrophy" incorporates an assortment of hereditary disorders that lead to progressive, generalized disease of the muscle prompted by inadequate or missing glycoproteins in the muscle cell plasma membrane. This activity outlines the evaluation and treatment of muscular dystrophy and highlights the role of the healthcare team in managing patients with this condition.

Objectives:

  • Identify the etiological related to muscular dystrophy.
  • Review the evaluation and functional assessment of various muscular dystrophies.
  • Outline the available management alternatives for muscle dystrophy.
  • Summarize interprofessional team strategies for improving care coordination and communication to improve outcomes for patients affected by muscular dystrophy.

Introduction

General Information

The term "muscular dystrophy" incorporates an assortment of hereditary disorders that lead to progressive, generalized disease of the muscle prompted by inadequate or missing glycoproteins in the muscle cell plasma membrane.[1] Muscular dystrophy is a non-communicable disorder with abundant variations.[2] Each has its pattern of inheritance, onset period, and the rate at which muscle is lost.[2] Alterations in specific genes cause different representations of this disease.

Anatomy 

Research has established that the gracilis, semimembranosus, semitendinosus, and sartorius muscles can be affected in patients with muscular dystrophy.[3] Feet can exhibit an equinovarus deformity.[4] The pelvis can tilt.[5] There may be contractures throughout the body.[6] Spinal deformities may produce lordosis or scoliosis.[7] The eye can exhibit cataracts and bilateral ptosis.[8]

Natural History

Onset usually occurs in the third to fourth decades.[9] But, it may reveal in infancy or undergo accelerated deterioration near the age of onset.[10] Parents of affected individuals may present concern that their child is not walking as well as other children their age.[11][12] The child may have trouble kicking a ball due to weakness.[13] Pseudo-drop events caused by weakness of quadriceps muscle may also be present.[5] Both parents could be healthy.[14] On physical examination, the affected individual will have massive calf muscles plus lower limb proximal muscle weakness.[15] This condition will make affected individuals want to utilize their arms and hands to aid in rising from a seated position.[16] Other complaints can involve a history of delayed ambulation, toe walking, calf hypertrophy, and proximal hip girdle muscle instability.[17] 

Presentations may also incorporate asymptomatic elevation of serum creatine kinase (CK), exertion intolerance, dilated cardiomyopathy, malignant hyperthermia, quadriceps myopathy, language delay, and Turner syndrome (Duchenne in X chromosome monozygotic females).[18] For some with subclinical muscular dystrophy, the diagnosis is initially suspected by family history or the appearance of raised liver enzymes, the basis for which is unclear.[19] These enzymes may include alanine aminotransferase and aspartate aminotransferase.[19] 

Since the inheritance of muscular dystrophy can be X-linked, the overwhelming majority of patients are male.[20] Symptomatic disease in daughters is explainable by Turner syndrome, skewed X chromosome inactivation, translocation of the mutated gene to an autosome, or uniparental disomy (both copies of a chromosome set originated of one parent).[21] Usually, symptomatic females present in infancy with proximal muscle weakness.[22] Reports exist of increased weakness in adulthood, myalgias, spasms, and lethargy as initial manifestations.[15] Scoliosis and sustained alveolar hypoventilation can cause severe problems for every child with muscular dystrophy.[23]

Patterns of Spread

Muscular dystrophy can be caused by mutations in numerous genes and can be transferred in an X-linked, autosomal dominant, or autosomal recessive fashion.[24] Changes in the X-linked gene DMD, which encodes dystrophin, is the most frequent cause of muscular dystrophy.[20] This why the phenotype is manifested in hemizygous males because they have only a single copy of the X chromosome.[25] One should note that mutations in dystrophin also create allelic heterogeneity.[26] Mutations in the DMD gene, for example, may cause muscular dystrophy of both Duchenne or the less serious Becker, based on the extent of the lack of protein.[27] 

Although the phenotypic characteristics of some of these disorders are definite, the phenotypic spectrum produced by mutations in various genes overlaps, whereby spanning to nonallelic heterogeneity.[28] Identification of nonallelic heterogeneity is critical for specific reasons: (1) the capacity to recognize disease loci in linkage studies is decreased by introducing subjects with associated phenotypes, but separate genetic disorders[29]; (2) genetic testing is further complicated because several distinct genes need to be analyzed along with the likelihood of distinctive mutations in all of the candidate genes[29]; and (3) data is concerned about how genes or proteins associate, therefore providing novel insights inside cell molecular physiology.[29] 

Phenocopies are thus produced, which are incidents in which nongenetic diseases simulate a genetic disorder. [30] For instance, features of virus or toxin-induced neurologic symptoms can mirror those seen in muscular dystrophy. As in nonallelic heterogeneity, phenocopies continue to confound linkage studies and genetic testing.[30] Patient history and accurate differentiation in phenotype can usually render signs that differentiate these maladies from similar genetic diseases. It is important to note that muscular dystrophy has variable expressivity and incomplete penetrance and, therefore, may be combined over a phenotypic spectrum in various affected individuals, further demonstrating the aspect of variable expressivity.[31]  

Etiology

Cause  

Muscular dystrophy most often results from defective or absent glycoproteins in the muscle membrane.[1] Each type of muscular dystrophy results from different gene deletions or mutations, causing various enzymatic or metabolic defects. [32] The dystrophin gene is the largest in the human genome, with 79 exons.[33] The dystrophin gene is subject to a high rate of spontaneous mutations because of its enormous size (>2 × 106 bases).[34]

Effect

Modes of Inheritance  

  • Autosomal Dominant, Autosomal Recessive, X-Linked: Emery-Dreifuss (EDMD)
  • Autosomal Dominant, Autosomal Recessive: Limb-Girdle (Dysferlinopathy, Erb), Pelvifemoral, Scapulohumeral
  • Autosomal Dominant: Facioscapulohumeral (Landouzy-Dejerine), Late-Onset Distal (>40 Years old), Myotonic, Oculopharyngeal, Scapuloperoneal
  • Autosomal Recessive: Congenital, Early Onset Distal (<40 years old)
  • X-Linked: Becker (Benign Pseudohypertrophic), Duchenne (pseudohypertrophic)

Autosomal Dominant, Autosomal Recessive, X-Linked Inheritance

  • Emery-Dreifuss (EDMD)

Emery-Dreifuss Muscular Dystrophy: Caused by an X-linked recessive defect in nuclear protein emerin at the Xq27-28 position.[35] This variant can also result from an autosomal recessive or autosomal dominant defect in inner nuclear lamina proteins lamin A/C on chromosome 1.[36]

Autosomal Dominant, Autosomal Recessive Inheritance

  • Limb-Girdle (Erb)
  • Pelvifemoral *
  • Scapulohumeral *

Limb-Girdle (Erb) Muscular Dystrophy: The majority are autosomal recessive but can be autosomal dominant.[37] The age of onset is variable with the distribution of involved muscles to include limbs and trunk. May display a heterogeneous phenotype.[37] The recessive form of the disease tends to have an earlier onset and progresses more quickly, whereas the dominant form follows a slower and more variable course.[24] Several different genes have been implicated in this disease—this type of muscular dystrophy correlates with deficiencies identified in multiple proteins. Sarcoglycan, calpain, dystroglycan, and dysferlin may be most common.[38] Also may involve telethonin, lamin A/C, myotilin, and caveolin-3.[39] LGMB 1A: is caused by myotilin gene deletion.[40] 

LGMB 1B results from by lamin A/C gene deletion.[41] LGMD 1C is the result of a caveolin 3 gene deletion.[42] A calpain gene mutation causes LGMD 2A.[43] LGMD 2B is caused by dysferlin gene deletion.[44] LGMD 2C is caused by sarcoglycan gene deletion.[45] LGMD 2D is caused by sarcoglycan gene deletion.[46] LGMD 2E is caused by sarcoglycan gene deletion.[47] LGMD 2F is caused by sarcoglycan gene deletion.[48] LGMD 2G results from telethonin gene mutation.[49] LGMD 2H is caused by a TRIM32 (tripartite motif-containing) gene 32 mutation.[50] LGMD 2I is the result of a fukutin-related protein gene deletion.[51]

Autosomal Dominant Inheritance

  • Facioscapulohumeral (Landouzy-Dejerine)
  • Late-Onset Distal (>40 Years old) *
  • Myotonic
  • Oculopharyngeal
  • Scapuloperoneal *

Facioscapulohumeral (FSHD) Muscular Dystrophy: Caused by an autosomal dominant deletion of 3.3 kb repeat on chromosome 4.[52] Approximately 95% of cases are due to a mutation in the D4Z4 region in FSHD1.[53] Other areas, such as the SMCHD1 region in FSHD2, can also cause this disease.[54] The age of onset is approximately 10 to 30 years old, with the distribution of affected muscles involved to be face, neck, and shoulders.[53]

Myotonic Muscular Dystrophy: Myotonic muscular dystrophy (or simply Myotonic dystrophy) results from the impaired expression of the Dystrophia Myotonica Protein Kinase (DMPK).[55] Caused by an autosomal dominant abnormally expanded CTG trinucleotide repeat sequence located in the 3′ untranslated region of the Dystrophia Myotonica Protein Kinase (DMPK) gene.[56] Because this mechanism involves the expansion of trinucleotide repeat sequences (CTG), the phenomenon of amplification and anticipation occurs (i.e., family members get the disease at earlier and earlier ages throughout the generations).[57] 

Clinical severity increases as the number of nucleotide repeats increases; some cases can be in the thousands.[58] Age of onset is approximately 10 to 15 years old with the distribution of involved muscles to include face and extremities. Some cases can be in the thousands.[58] This defect is classically associated with chromosome 19; however, a second form can occur on chromosome 3q.[10][59]

Oculopharyngeal Muscular Dystrophy: Age of onset is approximately 30 to 40 years of age. Distribution of involved muscles includes the extraocular and pharyngeal muscles. Caused by an autosomal dominant GCG trinucleotide repeat resulting in deficient mRNA transfer from the nucleus.[60]

Autosomal Recessive Inheritance

  • Congenital
  • Early Onset Distal (<40 years old) *

Congenital Muscular Dystrophy: Caused by a mutation of the sarcolemmal protein Merosin gene, deficiencies or mutations in laminin-alpha 2, collagen type VI, integrin-alpha 7, and glycosyltransferases.[61]

X-Linked Inheritance

  • Becker (benign pseudohypertrophic)
  • Duchenne (pseudohypertrophic)

Becker Muscular Dystrophy: Caused by a mutation of muscle protein dystrophin gene, which codes for the protein dystrophin, with 79 exons, by far the largest gene known in humans.[32] This gene transmitted in an X-linked re­cessive manner.[32] Its location is on the small arm (p) of the X chromosome at the Xp21 locus position.[62] Without dystrophin, muscle cells deteriorate or die. The age of onset is 10 to 20 years old, with the distribution of involved muscles to be generalized.[1] 

Duchenne Muscular Dystrophy: Caused by a mutation of the dystrophin gene, located on the small arm (p) of the X chromosome at the Xp21 position.[32] A spontaneous mutation occurs in a third of cases.[63][64] X-linked recessive maternal-fetal transmission occurs in the other two-thirds of cases.[63] The effect is resulting in a non-functional dystrophin protein, which causes similar effects as to that seen in Becker muscular dystrophy. Age of onset is approximately 3 to 5 years of age, with the distribution of involved muscles to be generalized.[61] 

Female Duchenne muscular dystrophy results from an error in female somatic cells whereby one X-chromosome becomes inactivated at an early stage, creating a mosaic representation of heterozygous X-linked genes. This condition generally is sufficient to protect female heterozygotes from X-linked disorders that affect males. However, X-inactivation in the female carrier of an X-autosome translocation can sometimes create a lethal genetic imbalance in half of the body's cells, causing those cells to die. The result is that the same X-chromosome expresses in every cell, and if that chromosome carries a disease allele, the individual can express the X-linked disease like a male, which explains some female cases of Duchenne muscular dystrophy.[65]

* A subtype or classification that is beyond the scope of the article but included for the sake of completeness. 

Epidemiology

  • Most Common Childhood Muscular Dystrophy: Duchenne.[38]
  • Most Common Adult Muscular Dystrophy: Myotonic.[9] 
  • Prevalence Of Muscular Dystrophy (General Population): 16 to 25.1 per 100,000.[66] [67] 
  • Frequency Of Muscular Dystrophy (General Population): 1 per 3,000 to 8,000.[68][69] 
  • Incidence Of Muscular Dystrophy (Male Births): 1 per 5,000 or 200 per 1,000,000.[70]

Becker Muscular Dystrophy 

  • Prevalence (General Population): 4.78 per 100,000.[71]
  • Frequency (Live Male Births): 1 per 18,000.[72] 
  • Incidence (Live Male Births): 1 per 5618.[72]

Congenital Muscular Dystrophy 

  • Prevalence (General Population): 0.99 per 100,000.[71] 
  • Prevalence (Children): 0.82 Per 100,000.[71] 
  • Prevalence (General Population-Italy): 1 per 16,000.[73] 
  • Incidence (General Population-Italy): 0.563 per 100,000.[74]

Duchenne Muscular Dystrophy 

  • Prevalence (General Population): 4.78 per 100,000.[71]  
  • Frequency (General Population): 13 to 33 per 100,000.[75] 
  • Frequency (Males): 1 per 3,500. [20] 
  • Incidence (Live Male Births): 1 per 5,136.[70] 

Emerry-Dreifuss Muscular Dystrophy

  • Prevalence (General Population): 0.39 per 100,000.[71] 
  • Prevalence (Children): 0.22 per 100,000.[71]

Facioscapulohumeral Muscular Dystrophy 

  • Prevalence (General Population): 3.95 per 100,000.[71]
  • Prevalence (Children): 0.29 per 100,000.[71]

Limb-Girdle (Erb) Muscular Dystrophy

  • Prevalence (General Population): 1.63 per 100,000.[71] 
  • Prevalence (Children): 0.48 per 100,000.[71]

Myotonic Muscular Dystrophy 

  • Prevalence (General Population): 8.26 per 100,000.[71] 
  • Prevalence (Children): 1.41 per 100,000.[71]

Oculopharyngeal Muscular Dystrophy 

  • Prevalence (General Population): 0.13 per 100,000.[76]

*US, England, Australia, & Canada unless otherwise specified* 

Pathophysiology

Description

  • Muscle contraction
  • Dystrophin
  • Dystrophin-glycoprotein complex

Muscle Contraction: It is essential first to understand the underlying physiology of muscle cell function. The sliding filament model represents muscle tension as a function that depends on the contraction of the muscle filaments; this is promoted by calcium, which, delivered from the sarcoplasmic reticulum, leads to muscle depolarization. Intracellular calcium binds to the anionic charge of troponin C, which leads to tropomyosin uprooting off of the G-actin site. Once exposed, a myosin head attaches to the presented G-actin site generating a pivot that requires energy in the form of ATP (adenosine triphosphate) to function. This pivot lets filaments made of actin to slide past filaments made of myosin, which achieves muscle shortening, which transfers to the muscle cell's glycoprotein rich cytoskeleton.

Dystrophin: Dystrophin is restricted to the cytoplasmic surface of the muscle fiber plasma membrane and connects the internal cytoskeleton to the extracellular matrix via glycoproteins that traverse the plasma membrane.[32] This cytoskeletal protein renders structural stability to a protein (dystroglycan) complex in cell membranes.[77] More precisely, dystrophin is what anchors the actin cytoskeleton to the basement membrane within a membrane-glycoprotein complex.[77] Dystrophin, laminin, and other proteins are a part of this cytoskeletal framework.[32] Dystrophin connects with F-actin and with β-dystroglycan, which then attaches to α-dystroglycan and laminin within the extracellular matrix (ECM).[32][78] Dystrophin's role is to secure the cytoskeleton to the extracellular matrix.[32] Consequently, if dystrophin is not performing correctly, this modifies tension transmission in a contracting muscle.[1] The contractile actin and myosin proteins normally shorten, which sequentially issues toward both muscle weakness and injury to the cell membrane. Creatine kinase suddenly oozes from every damaged muscle cell and consequently is found in abnormally high levels in the plasma.[79] This CK release additionally incites an inflammatory response that promotes scar tissue formation ending in the classic pseudohypertrophy of the calf muscles linked with muscular dystrophy.[80] Even though the muscles look hypertrophied, there exists a deficiency in working contractile filaments in the tissue and therefore creates weak muscles.[1] The­ deficit is present from fetal development onwards.[81] Phago­cytosis of the damaged muscle cells by inflammatory cells cause scar­ring and further impairment of muscle function.[1]

Dystrophin-Glycoprotein Complex: This protein meshwork seems to strengthen the sarcolemma.[82] The loss of one part of the network may generate changes in other elements. For instance, an initial loss of dystrophin may drive the destruction of sarcoglycans, including dystroglycan.[83] The weakening of the membrane leads to muscle cell death.[1] Skeletal muscle comes to be almost completely substituted with fat and connective tissue.[84] The skeleton even­tually becomes distorted, inducing gradual immobility. Car­diac muscle and smooth muscle of the gastrointestinal tract typically be­come fibrotic.[85] The brain manifests structural deformities with no apparent consistency.[85]

Most Common Findings

  • Chronic respiratory infections precipitated by weakness in the smooth muscle of bronchioles.[23]
  • Impotence caused by gonadal atrophy, which is characteristically associated with myotonic dystrophy.[86] 
  • It is common to possess dysphagia, which is esophageal muscle involvement.[87]
  • Myotonia is a term that describes the inability to relax muscles, which classically manifests as an inability to loosen one's grip or release a handshake.[17] 
  • As a pediatric disease, parents will often complain that their child is clumsy or becomes extremely weak quickly.[88] 
  • Gower sign is when subjects try to stand from a supine position, they "march" their hands and feet to each other).[89] 
  • Weakness and stiffness of distal muscles are usually the presenting symptoms in adolescents with myotonic dystrophy.[15] 

Toxicokinetics

Malignant Hyperthermia: This is a unique and life-threatening myopathy that can occur in genetically susceptive individuals following exposure to triggering factors, usually halogenated anesthetic gases like halothane, isoflurane, sevoflurane, and desflurane.[90] This condition can also manifest from exposure to succinylcholine.[91] These, when given to susceptive individuals, may drive toward extreme skeletal muscle contraction.

Patients with dystrophinopathies produce rhabdomyolysis if exposed to those agents, and many scholars consider it reasonable to utilize non-triggering agents exclusively. Malignant hyperthermia happens because of the irregular calcium control correlated with dytrohpinopathies.

Dysregulated calcium release of the sarcoplasmic reticulum mixed with anesthetic-induced interference of calcium reuptake ends in constant muscle contraction, prolonged aerobic and anaerobic metabolism, driving up carbon dioxide production, elevated end-tidal carbon dioxide, and tachycardia in these individuals who are unable to raise their alveolar minute volume properly. This hypermetabolism generates an exponentially dangerous amount of heat. So much so that the amount of heat appearing overwhelms the body's capacity to dissipate this heat.

History and Physical

Cardiovascular Findings

  • Arrhythmias
  • Congestive heart failure
  • Dilated cardiomyopathy 

Arrhythmias: Cardiac arrhythmias are even more significant in patients with muscular dystrophy. The typical electrocardiogram (ECG) shows increased net RS in lead V1; deep, narrow Q waves from the precordial leads, with tall right precordial R waves in V1.[92] Cardiac disturbances occur commonly in patients with Duchenne muscular dystrophy Type 1 (DM1).[93] Myotonic dystrophy affects the heart muscle, causing arrhythmias and heart block.[94] ECG abnormalities include first-degree heart block and more extensive conduction system involvement. A complete heart block and sudden death can occur.[95]

Congestive Heart Failure: Congestive heart failure seldom occurs except with severe stress, such as pneumonia.[96] Congestive heart failure occurs infrequently but may result from cor pulmonale secondary to respiratory failure.[97] Mitral valve prolapse also occurs commonly.[97]

Dilated Cardiomyopathy: Genetic dilated cardiomyopathies account for 30 to 40% of cases of nonischemic dilated cardiomyopathies.[98] Some are associated with muscular dystrophy. In the skeletal myopathies, a dominant R wave in lead V1 (indicative of prominent posterior wall involvement, by the same mechanism as in posterior wall myocardial infarction).[99] A cardiac cause of death is not always certain despite the presence of cardiomyopathy in almost all patients.[100] The incidence of cardiac involvement in Duchenne muscular dystrophy is as high as 95%.[101] Chronic heart failure may occur in 50% of children.[102]

Musculoskeletal Findings

  • Contractures
  • Delayed motor milestones
  • Expressionless Facies
  • Fractures
  • Gait instability 
  • Gower's sign
  • Muscle wasting
  • Myotonia
  • Pseudohypertrophy
  • Proximal muscle weakness
  • Scoliosis
  • Toe walking

Contractures: Most patients have joint contractures of varying degrees at elbows, hips, knees, and ankles. Contractures that present at birth are referred to as arthrogryposis. Contractures of both the heel cords and iliotibial bands manifest by age six years when toe walking is associated with a lordotic posture. joint contractures and limitations of hip flexion and extension of the knee, elbow, and wrist are made worse by prolonged sitting.[103] Contractures become fixed, and progressive scoliosis often develops that may be associated with pain.[103] Contractures and muscle wasting contribute to muscular atrophy and deformity of the skeleton. Duchenne muscular dystrophy has serious complications.[24] Muscle weakness also leads to contractures of the knees, hips, and other joints, and scoliosis develops in most boys with Duchenne disease.[104] The contractures and skeletal deformities that develop from facioscapulohumeral muscular dystrophy are less often and are less prominent than in Duchenne muscular dystrophy.[105] Other clinical findings include brought about by a futile attempt to overcome foot drop and depressed or absent muscle stretch reflexes.[106]

Delayed Motor Milestones: Duchenne muscular dystrophy is usually identified in children at approximately three years of age when the parents first notice slow motor development.[107] Clinical symptoms often begin between 5 and 15 years of age.[107] Sitting, standing, and walking are developmentally delayed, and the child is clumsy, falls frequently, and has difficulty climbing stairs.[107] Children with Becker muscular dystrophy remain ambulatory into their teens and early 20s; in one study, the average age at the time of necessity for a wheelchair was 25 years.[17] By definition, patients with Becker dystrophy can walk beyond age 15, whereas patients with Duchenne dystrophy typically are wheelchair-bound by age 12.[107]

Expressionless Facies: The inability to close the eyes completely may be noted from early childhood. The face is expressionless, and the pouting of the lips makes whistling impossible. The muscular weakness and wasting produce a" drooping expression." In myotonic dystrophy, the face is hatchet-shaped due to the facial wasting and weakness, and there is bilateral partial ptosis. With bilateral facial palsy, although very rare, it is important to rule out all other possible diagnoses for facial weakness, including upper motor neuron and lower motor neuron disorders, with thorough diagnostic tests.[108]

Fractures: Muscle weakness and inactivity, particularly once a person is in a wheelchair full time, lead to osteoporosis and pathologic fractures. If a fracture occurs, bisphosphonates may help to strengthen bone, although there are no long-term studies on safety in this population.[109]

Gait Instability: The boys stumble repeatedly and have trouble keeping up with friends when playing. Running, jumping, and hopping is always abnormal.[110]

Gower's Sign: This clinical sign can be evoked by asking the child to stand from a sitting position.[111]  Children with muscular dystrophy and other disorders with muscle wasting will not possess the muscle force to stand.[111] They may alternatively first move into a prone position, thrust themselves onto all fours, and suddenly "walk" their hands along their thighs to a standing posture.[111] The appearance of a Gower sign signifies significant proximal muscle weakness. More specifically, it is caused by the weakness of the lumbar and gluteal muscles.[111]

Muscle Wasting: Muscular weakness always begins in the pelvic girdle, causing a "waddling" gait. [107] Hypertrophy of the calf muscles is apparent in 80% of cases.[112] The pattern of muscle wasting present in Becker muscular dystrophy bears a close resemblance to that seen in Duchenne disease. Proximal muscles, particularly of the lower extremities, are prominently involved. As the disease progresses, weakness becomes more generalized.[113] Duchenne weakness worsens over the subsequent few years, resulting in the loss of ability to ambulate by 8 to 13 years of age. Myotonic dystrophy patients have a common "hatchet-faced" look due to temporalis, masseter, and facial muscle atrophy.

Myotonia: TThe word myotonia applies to a prolonged unconscious muscle contraction demonstrated through not being able to loosen grasp. The patient may have a delay in releasing grip when shaking hands. A pause in opening and closing the fists is observable. Myotonia emerges typically at the age of 5 years and is demonstrable through percussion of the thenar eminence, jaw, and musculature of the forearm. Upon forced voluntary closures, myotonia triggers a sluggish relaxation. As muscle deterioration progresses, so does the difficulty of detecting myotonia. It is difficult for people with myotonic dystrophy to relax their grip, in particular whenever the subject is cold.[109][114] 

Pseudohypertrophy: Pseudohypertrophy of the muscle extends to the toes.[115]  Boys will exhibit, in preschool, with muscle weakness, trouble walking, and wide calves (pseudohypertrophy) caused by healthy muscle fiber replacement with fat and connective tissue.[116]  Although the calves are big, the muscle is small. Enlargement of muscles, particularly in the calves, is an early and prominent finding. Congenital can also have calf hypertrophy.[117]

Proximal Muscle Weakness: Proximal muscle weakness may be remarkably apparent when contrasted to distal weakness in many of the conditions listed.[118] Specific evaluation for this, such as arising from a low seat or a squatting posture, is needed.[24] In 3 to 5 years, muscles of the shoulder girdle become affected in Duchenne. This deterioration is succeeded with bilateral sternocleidomastoid and trapezius; myalgias without weakness; winging of the scapula; continuous muscle fiber loss leading to weakness principally of the voluntary muscles; proximal arms and legs. Congenital dystrophy forms may present with hypotonia plus proximal or generalized muscle weakness. Loss of muscle strength is progressive, with leg involvement more severe than arm involvement. 

Between 8 and 10 years of age, walking may require the use of braces. Clinical instability originates in the pelvic girdle, first generating difficulty standing from the floor (Gower sign), climbing stairs, and a waddling gait as a result of the weakness in the lumbar and gluteal muscles.[17] Facioscapulohumeral muscular dystrophy, as the name implies, begins with weakness and atrophy of facial and shoulder girdle (scapulohumeral) muscles.[119] The diagnosis of limb-girdle muscular dystrophy is accepted with the elimination of acute events causing proximal weakness, and the clinical picture, including genetic pattern, excludes Duchenne and facioscapulohumeral muscular dystrophy.[119] Neck muscles, including flexors, sternocleidomastoids, and distal limb muscles, are compromised early in myotonic dystrophy.[17] The weakness of wrist extensors, finger extensors, and intrinsic hand muscles impairs function in myotonic dystrophy.[17] Ankle dorsiflexor weakness may induce a foot drop in myotonic dystrophy.[17] 

Interestingly, contrary to the mechanism of weakness, proximal muscles continue to become more powerful throughout the course in myotonic dystrophy. There is, however, preferential atrophy and weakness of quadriceps muscles in myotonic dystrophy.[17] Scapuloperoneal muscular dystrophy is considered a variant of facioscapulohumeral muscular dystrophy. Their distinction is that the distal muscles in the lower extremity are involved early instead of the facial and shoulder muscle weakness, which is the early sign in facioscapulohumeral dystrophy.[17]

Scoliosis: Once scoliosis begins, it is relentlessly progressive. Curves of more than 20 degrees require surgical intervention to maintain pulmonary function.[120]

Toe Walking: The foot assumes an equinovarus position, and the child tends to walk on the toes because of the weakness of the anterior tibial and peroneal muscles.[121] Patients with muscular dystrophy often toe-walk because of the weakness of the anterior tibial and peroneal muscles, causing the feet to assume a talipes equinovarus position.[122] 

Neurological Findings

  • Cognitive dysfunction
  • Hypersomnia
  • Seizures
  • Visual disturbances

Cognitive Dysfunction: Mild to moderate cognitive problems are common but not universal.[123] Intellectual impairment in Duchenne dystrophy is common; the average intelligence quotient is approximately one standard deviation below the mean.[124] A moderate degree of intellectual disability causes these children to have a mean IQ of approximately 80.[125] Impairment of intellectual function appears to be nonprogressive and affects verbal ability more than performance.[126] Mental retardation may occur in Becker dystrophy, but it is not as common as in Duchenne. The central nervous system is affected in some forms of congenital muscular dystrophy.[127][123]

Hypersomnia: The excessive urge to sleep and daytime somnolence is common.[128]

Seizures: In merosin and FKRP deficiency, only a small number of patients have mental retardation and seizures.[129]

Visual Disturbances: Ocular abnormalities due to eye muscle dysfunction lead to impaired vision.[130] Myotonic dystrophy may present as difficulty in opening eyes after tight closure.[131]

Other Findings

  • Bladder instability
  • Cataracts
  • Frontal baldness
  • Generalized digestive complaints
  • Gonadal atrophy and hypogonadism
  • Insulin resistance

Bladder Instability: Bladder functions are often mildly affected with urinary urgency as a frequent symptom.[132] 

Cataracts: The affected individual may be referred from an ophthalmologist after a recent examination, which revealed the potential for an underlying disorder such as myotonic dystrophy (iridescent spots).[133][134]

Frontal Baldness: This is characteristic of myotonic dystrophy, possibly due to gonadal atrophy and subsequent hypogonadism.[9]

Generalized Digestive Complaints: Smooth muscle dysfunction may cause megacolon, volvulus, cramping pain, and malabsorption in the gastrointestinal tract.[135] Disturbed gastrointestinal peristalsis. Decreased esophageal and colonic motility.[136] Bowel functions are often mildly affected with constipation as a frequent symptom.[137] Some causes of death include aspiration of food and acute gastric dilation. Palatal, pharyngeal, and tongue involvement produce a dysarthric speech, nasal voice, and swallowing problems.[138][139]

Gonadal Atrophy and Hypogonadism: Small soft testes suggesting atrophy are associated with myotonic dystrophy leading to hypogonadism.[140][9]

Insulin Resistance: Diabetes is commonly associated with muscular dystrophy.[141] 

Respiratory Findings

  • Chest deformity
  • Recurrent pulmonary infections
  • Respiratory insufficiency
  • Sleep apnea

Chest Deformity: The chest deformity with scoliosis impairs pulmonary function, which is already diminished by muscle weakness.[142] Examine for gynecomastia, which can be present in patients with myotonic dystrophy. 

Recurrent Pulmonary Infections: By age 16 to 18 years, patients are predisposed to serious, sometimes fatal pulmonary infections.[143] Susceptibility to respiratory tract infections and progressive deterioration of pulmonary function generally lead to premature death, usually into the twenties.[144]

Respiratory Insufficiency: Respiratory failure is the commonest cause of death.[23] Inspection will identify the areas of muscle wasting. A patient presenting with rapid-onset muscle weakness requires an urgent full assessment, as respiratory muscle involvement may lead to respiratory failure.[145] Some patients have a diaphragm and intercostal muscle weakness, resulting in respiratory insufficiency.[10] Progressive course resulting in respiratory complications. Some cases lead to respiratory failure. Pulmonary function is significantly compromised because of marked kyphoscoliosis ("humped" upper spine combined with scoliosis), which usually develops once the child is confined to a wheelchair. As children age, muscle weakness progresses, and respiratory weakness leads to breathing difficulty, particularly when sleeping.[146]

Sleep Apnea: An increased need or desire for sleep is common, as is diminished motivation.[147]

Evaluation

Laboratory Tests

  • Alanine aminotransferase (ALT, SGPT)
  • Aldolase (serum)
  • Arterial blood gases (ABG)
  • Aspartate aminotransferase (AST)
  • Creatine kinase (CK, CPK) and creatine kinase isoenzymes (CK-MB & CK-MM)
  • Lactate dehydrogenase (LDH)
  • Urinalysis (UA)

Alanine Aminotransferase (ALT, SGPT): The normal range in males is 10 to 40 U/L. The normal range in females is 8 to 35 U/L; it is elevated in muscular dystrophy.[19]

Aldolase (Serum): The normal range is 0 to 6 U/L. It is elevated in muscular dystrophy but decreases in later stages of muscular dystrophy.[148]

Arterial Blood Gases (ABG): Normal ranges: PO2 is 75 to 100 mmHg; PCO2 is 35 to 45 mm Hg; HCO3- is 24 to 28 mEq/L; pH is 7.35 to 7.45. Respiratory acidosis can develop if there are defects in muscles involved in respiration.[149]

Aspartate Aminotransferase (AST): Normal ranges from 0 to 35 U/L. Elevated in muscular dystrophy.[19]

Creatine Kinase (CK, CPK) and Creatine Kinase Isoenzymes (CK-MB and CK-MM): Normal ranges from 0 to 130 U/L. Elevated in muscular dystrophy (hyperCKemia).[150] The serum enzymes, especially creatine phosphokinase (CPK), are increased to more than ten times normal, even in infancy and before the onset of weakness.[151] Diagnosis is suggested (a high creatine kinase [CK] level does not confirm the diagnosis because many other alterations can also increase CK) by measuring the blood creatine kinase level, which can be 100 times the normal level, with diagnostic confirmation by genetic testing for mutations in the dystrophin gene.[152] Serum CK levels are invariably elevated between 20 and 100 times normal in Duchenne muscular dystrophy.[153] The levels are abnormal at birth, but values decline late in the disease because of inactivity and loss of muscle mass.[154] Elevated CPK levels at birth are diagnostic indicators of Duchenne muscular dystrophy.[155] The identification of female carriers of the disease is not achievable with certainty, but serum CPK is elevated in 60% to 80% of carriers.[156] Serum CK can be 2- to 20 times above normal in Emery-Dreifuss muscular dystrophy.[157] Myotonic dystrophy may be associated with a normal CK or only mild elevation.[93]

Lactate Dehydrogenase (LDH): Normal ranges from 50 to 150 U/L. Elevated in muscular dystrophy. LDH 4: 3 to 10%, LDH 5: 2 to 9%.[148]

Urinalysis (UA): Glucose in urine is commonly associated with muscular dystrophy due to the high incidence of diabetes mellitus within this population.[158] Myoglobinuria may also be present.[148]

Radiographic Tests 

  • Magnetic Resonance Imaging (MRI)
  • Computerized Tomography (CT)

Magnetic Resonance Imaging (MRI): Coronal T1 weighted MRI may confirm the nonuniform fatty atrophy.[159] There will be a relatively normal sartorius. Lateral radiographs may show cavus foot deformity and diffuse osteopenia.[160] The sagittal view will show diffuse fat replacement of the gastrocnemius & semimembranosus muscles. These changes contribute to the prominent calves typical of affected children.[116]

Computerized Tomography (CT): Axial CT shows denervation hypertrophy of the tensor fascia lata. The muscle becomes enlarged with an increase in intramuscular fat.[161]

Other Tests

  • Chromosomal analysis
  • Electrocardiogram (ECG)
  • Electromyography (EMG)
  • Genetic testing
  • Immunocytochemistry
  • Muscle biopsy
  • Polysomnogram
  • Slit lamp
  • Western blot analysis 

Chromosomal Analysis: DNA testing for common mutations and chromosomal analysis can now rule out Down syndrome, myotonic dystrophy, and other disorders. In both Becker and Duchenne dystrophies, the DNA deletion size does not predict clinical severity.[162] DNA deletion, however, does not change the translational read frame of the messenger RNA in approximately 95 percent of patients suffering from Becker dystrophy. Such "in-frame" mutations allow for the development of dystrophin, which explains for the existence of dystrophin on Western blot examination.[163]

Electrocardiogram (ECG): Often, patients will have annual echocardiograms to stay ahead of any developing cardiomyopathy. This study will demonstrate atrial and atrioventricular rhythm disturbances. The typical electrocardiogram shows an increased net RS in lead V1; deep, narrow Q waves in the precordial leads.[164] A QRS complex too narrow to be right bundle branch block; and tall right precordial R waves in V1.[165] Dominant R wave in lead V1 is the best clue to the actual diagnosis.[166] Normal PR interval, QRS duration. There are abnormal Q waves in the precordial leads. Prominent Q waves lead II.[167]  QRS axis shows deviation.[168] Significant ECG irregularities and diagnosis of atrial tachyarrhythmia are indicators of sudden death.[169] P wave with a prominent early deflection in lead V1 reflects right atrial enlargement.[167]  The ECG could mimic pulmonary hypertension evidenced most notably in lead V1 with QRS right axis deviation.[167] Another less likely cause is an old true posterior wall myocardial infarction. Therefore look for an associated inferior wall myocardial infarction, which will be absent if associated with the dystrophinopathies.[99] The increasing severity of conduction disease is indicated by increasing PR interval and QRS duration.[170] An implantable cardioverter-defibrillator should be considered in all patients.[95]

Electromyography (EMG): Allows assessment for denervation of muscle, myopathies, and myotonic dystrophy, motor neuron disease. EMG demonstrates features typical of myopathy.[171] Clinical examination, electromyography changes are found in almost any muscle: waxing and waning of potentials termed the dive bomber effect.[172]

Genetic Testing: A definitive diagnosis of muscular dystrophy can be established with mutation analysis on peripheral blood leukocytes.[173] Genetic testing demonstrates deletions or duplications of the dystrophin gene in 65% of patients with Becker dystrophy, which is approximately the same percentage as in Duchenne dystrophy.[174] Deletions are usually detected by multiplex polymerase chain reaction (PCR), other mutations by sequencing.[175] Carrier females may be identified by mutation testing or by linkage to intragenic markers allowing for an intragenic recombination rate of 12%.[176]

Immunocytochemistry: A definitive diagnosis of muscular dystrophy can be established based on dystrophin deficiency in a biopsy of muscle tissue.[177] Also, staining of muscle with dystrophin antibodies can demonstrate the absence or deficiency of dystrophin localizing to the sarcolemmal membrane.[1] DIsease carriers may demonstrate a mosaic pattern, but dystrophin analysis of muscle biopsy specimens for carrier detection is not reliable.[178] Immunohistochemistry reveals the absence of emerin staining of myonuclei in X-linked Emery-Dreifuss due to emerin mutations.[179]

Muscle Biopsy: The muscle biopsy shows muscle fibers of varying sizes as well as small groups of necrotic and regenerating fibers. Connective tissue and fat replace lost muscle fibers.  Muscle biopsy usually shows nonspecific dystrophic features, although cases associated with FHL1 mutations have features of myofibrillar myopathy.[180] Muscle biopsy shows muscle atrophy involving Type 1 fibers selectively in 50 percent of cases.[15] Typically various agentic nuclei can be observed in muscle cells as well as in dysplastic fibers with intracytoplasmic nuclear clusters.[181] Histologic changes in muscle include degeneration of muscle fibers, with variation in fiber size and central nuclei.[182] The individual with Scapuloperoneal muscular dystrophy will have fiber splitting and fibers that appear profusely "moth-eaten" and whorled.[183]

Polysomnogram: Excessive daytime somnolence with or without sleep apnea is not uncommon. Sleep studies, noninvasive respiratory support (biphasic positive airway pressure [BiPAP]), and treatment with modafinil may be beneficial.[184]

Slit Lamp: An examination for cataracts that may be present in patients with muscular dystrophy.[185]

Western Blot: A diagnosis of Duchenne dystrophy can also be made by Western blot analysis of muscle biopsy specimens, revealing abnormalities on the quantity and molecular weight of dystrophin protein. On Western blot, Becker muscular dystrophy individuals dystrophin levels will appear normal, although the protein itself is abnormal; this is in comparison to Duchenne muscular dystrophy affected individuals who have a significantly decreased dystrophin on Western blot.[186]

Treatment / Management

Medical Interventions

  • Anti-arrhythmics
  • Anti-epileptics
  • Anti-myotonic drugs
  • Non-steroidal anti-inflammatory drugs (NSAIDs)
  • Steroids

Anti-Arrhythmics: The pharmacological treatment of patients with a prevalent involvement of the cardiac tissue conduction relies on the use of ACE-inhibitors and appropriate antiarrhythmic drugs. In the case of atrial arrhythmias, the preference is for drugs such as antiarrhythmics (flecainide, propafenone) and beta-blockers. Amiodarone should be limited to patients who do not respond to the previous drugs, taking in mind that these are young patients, long-term therapy, and a high risk of adverse effects on the thyroid and pulmonary function.[187]

Anti-Epileptics: Children need to be followed closely by neurologists. Management of epilepsy is necessary for some patients.[188]

Anti-Myotonics: The pain associated with muscle rigidity is greatly alarming in the patient.[189] When myotonia is disabling, treatment with a sodium channel blocker such as phenytoin (100 mg orally three times daily), procainamide (0.5–1 g orally four times daily), or mexiletine (150 to 200 mg orally three times daily) may prove helpful, but the associated side effects, particularly for antiarrhythmic medications, are often limiting.[190] The preferred drug for a symptomatic patient requiring anti-myotonia medication is phenytoin and mexiletine; other medications, especially quinine and procainamide, can cause an increase in the risk of cardiovascular complications.[191]

Non-Steroidal Anti-Inflammatory Drugs: Treatment involves the administration of non-steroidal anti-inflammatory drugs to decrease pain and inflammation.[192]

Steroids: Glucocorticoids, administered as prednisone in a dose of 0.75 mg/kg per day, significantly slow progression of muscular dystrophy for up to 3 years.[193] Some patients cannot tolerate glucocorticoid therapy; weight gain and increased risk of fractures, in particular, represent a significant deterrent.[194] There is recent evidence that oral steroids early in the disease can lead to dramatically improved outcomes.[195] Children can walk an additional 2 to 5 years, and life expectancy has increased.[12] Researchers have not adequately studied the use of glucocorticoids in Becker dystrophy.[196] Some individuals with facioscapulohumeral muscular dystrophy improve with steroid therapy, particularly if the clinical picture includes rapidly progressive weakness.[197] The evidence is not clear on if steroids are useful for the treatment of myotonic muscular dystrophy.[198]

Golodirsen (SRP-4053): This drug is an antisense therapy used for the treatment of Duchenne muscular dystrophy. Patients need to have a confirmed mutation of the dystrophin gene to facilitate exon 53 skipping. It is FDA approved, but the evidence to support its use is not yet well established.[199][200]

Surgical Interventions

  • Defibrillator or a cardiac pacemaker
  • Contracture release
  • Shoulder surgery
  • Spinal correction

Contracture Release: Surgical release of contracture deformities is used to maintain normal function as long as possible.[201] Massage and heat treatments also may be helpful.

Defibrillator or Cardiac Pacemaker: Cardiac function requires monitoring, and pacemaker placement may be a consideration if there is evidence of heart block.[95] Management of cardiomyopathy and arrhythmias may be life-saving.[95] In patients with severe syncope, established conduction system disorders with second-degree heart block previously documented, or tri-fascicular conduction abnormalities with significant PR interval lengthening, consideration needs to be given towards placement of a cardiac pacemaker.[202] An advanced cardiac block is also an indication to install a pacemaker.[203]

Shoulder Surgery: Individuals with facioscapulohumeral muscular dystrophy may benefit from surgery to stabilize the shoulder.[204]

Spinal Correction: Scoliotic surgery is an option when curves exceed 20 degrees to prolong respiratory function or walking ability or both.[120]

Other Interventions

  • Supportive physiotherapy 
  • Supportive bracing
  • Supportive counseling
  • Genetic counseling

In addition to pharmacologic intervention, general supportive care may be beneficial, including physical therapy, range of motion exercises, padding, skincare, orthotics, and safety awareness.

Supportive Physiotherapy: Treatment includes supportive physiotherapy to prevent contractures and prolong ambulation.[64] Maintaining function in unaffected muscle groups for as long as possible is the primary goal. Although activity fosters maintenance of muscle function, strenuous exercise may hasten the breakdown of muscle fibers.[205]

Supportive Bracing: This helps to maintain normal function as long as possible  Proper wheelchair seating is essential. Molded ankle-foot orthoses help stabilize gait in patients with foot drop.[206] Lightweight plastic ankle-foot orthoses (AFOs) for footdrop are extremely helpful. Footdrop is easily treatable with AFOs.[207] Individuals with scapuloperoneal muscular dystrophy remain ambulatory for 40 or more years.[208]  Occasionally, walking may become hampered by paraspinal muscle contractures; in that case, a wheelchair may assist the individual when it is necessary to cover long distances.[103] Bracing may be performed for function; for example, dorsiflexion of the feet with ankle-foot orthotics to prevent tripping or to provide support and comfort.[103]

Supportive Counseling: Some forms of muscular dystrophy may be arrested for prolonged periods, and most patients remain active with a normal life expectancy.[209] Thus, vocational training and supportive counseling are important to provide the information necessary to plan their future.

Genetic Counseling — Genetic counseling is recommended.[210] With X-linked inheritance, male siblings of an affected child have a 50% chance of being affected, and female siblings have a 50% chance of being carriers. If the affected individual marries and has children, all daughters will be carriers of this X-linked recessive disorder. Genetic counseling should be offered to the mother, female siblings, offspring, and any maternal relatives.

Differential Diagnosis

Differential Diagnosis For Associated Signs And Symptoms

Utilizing the following mnemonics will aid in the recall of the differential diagnoses associated with the weakness and ataxia of muscular dystrophy:

Ataxia (Can't Stand Very Well)

  • Cerebellar Ataxia
  • Sensory Ataxia
  • Vestibular Ataxia
  • Weakness

Ataxia, Acute (U.N.A.B.L.E. T.O. S.T.A.N.D.)

  • Underlying weakness (may mimic ataxia)
  • Nutritional neuropathy (vitamin B12 deficiency)
  • Arteritis/vasculitis
  • Basilar migraine
  • Labyrinthitis/vestibular neuronitis
  • Encephalitis/infection
  • Trauma (postconcussive)
  • Other (rare metabolic or genetic diseases)
  • Stroke (ischemia or hemorrhage)
  • Toxins (drugs, toluene, mercury)
  • Alcohol Intoxication
  • Neoplasm/paraneoplastic syndromes
  • Demyelination (Miller Fisher, Guillain Barre, MS)

Ataxia, Chronic (C.A.N.T. S.T.A.N.D.)

  • Congenital malformation/Chiari
  • Autosomal recessive ataxias
  • Nutritional (vitamin B12 deficiency)
  • Trauma (postconcussive)
  • Stroke (ischemia or hemorrhage)
  • Toxins (drugs, toluene, mercury)
  • Alcohol Intoxication/Autosomal dominant ataxia
  • Neoplasm/paraneoplastic syndromes
  • Demyelination (MS)

Weakness, Acute (M.I.S.S. G.I.M.P)

  • Myelopathy (acute) / myopathy
  • Infection
  • Stroke
  • Systemic illness
  • Guillain-Barre syndrome
  • Iatrogenic/drugs
  • Myasthenia
  • Paralytic toxins/periodic paralysis

Weakness, Chronic (G, I.' A.M. L.I.M.P, C.A.N.T. S.T.A.N.D.)

  • Guillain-Barré syndrome
  • Iatrogenic (paralytic agents, aminoglycosides, steroids)
  • Myopathy/myositis
  • Lou Gehrig disease (ALS, motor neuron disease-usually gradual)
  • Infection (polio, botulism)
  • Myelopathy (acute).
  • Periodic paralysis, porphyria, paraproteinemia
  • Cushing's
  • Arteritis/vasculitis/stroke
  • Neoplastic (meningitis, paraneoplastic)
  • Toxins (lead, arsenic, pufferfish, tick paralysis)
  • * Acute/subacute, bilateral with minimal sensory involvement
  • Systemic illness (anemia)
  • Thyroid
  • Addison's
  • Neuromuscular junction disease (Myasthenia, Lambert-Eaton syndrome)
  • Diabetic amyotrophy 

Diseases Or Conditions Potentially Mistaken For This Disease

  • Acid maltase deficiency
  • Drug-induced myopathy
  • Dermatomyositis sine dermatitis
  • Endocrinopathy
  • "Fatigue" syndromes
  • Inclusion body myositis
  • Late-onset spinal muscular atrophy
  • McArdle's deficiency (myophosphorylase deficiency)
  • Metabolic myopathies
  • Motor neuron disease
  • Neurogenic disorders
  • Polymyositis

Diseases Or Conditions That Must Be Ruled Out

While muscular dystrophy is not a diagnosis of exclusion, it is often confused for several diseases due to their overlapping etiologies. The clinician must exclude the following diseases in a patient with a muscular disorder, which can otherwise cause significant morbidity and mortality:

  • Adrenal insufficiency
  • Electrolyte imbalance: sodium, potassium, and magnesium
  • Hypercalcemia
  • Porphyrias
  • Rabies
  • Complicated migraine
  • Postictal (Todd's) paralysis
  • Hypoglycemia

Acute spastic paraparesis is a medical emergency. The clinician should strongly suspect myasthenia gravis in any patient who reports excessive weakness at the end of the day. Consider Pancoast tumor of the lung in a patient presenting with upper limb weakness and painful neuropathy secondary to neoplastic infiltration of the lower trunk of the brachial plexus, particularly when Horner syndrome is present. Paraneoplastic syndromes must also merit consideration in the diagnosis. Rule out lesions of extrapyramidal tracts or cerebellar pathways which may also present as weakness, but without objective evidence of decreased muscle strength.

An acute onset gait disorder is likely to be due to acute systemic decompensation; a careful, systematic evaluation can exclude such catastrophic presentations. It is not advisable to attribute gait disorder to one single disease. An abnormal gait may be a risk factor for falls. Difficulty with getting out of a chair without arm support and initiate movement suggests limb-girdle dystrophy. 

Gait abnormalities can indicate a serious medical emergency, especially when the problem is associated with any of these additional symptoms:

  • Headache (raised intracranial pressure)
  • Nausea or vomiting
  • Decreased alertness
  • Impaired coordination presenting on only one side of the body
  • Recent viral illness/immunization (Guillain-Barré syndrome)
  • Trauma.

Discussion 

Muscular dystrophy may be mistaken for polymyositis, most often when the duration of symptoms appears to be short. The pattern of weakness may be very helpful in making a distinction. Secondary inflammatory infiltrates may cause pathological confusion, especially in limb-girdle muscular dystrophy and facioscapulohumeral muscular dystrophy.

Are Reflexes Hypoactive? The presence of hypoactive reflexes would suggest myotonic dystrophy, tabes dorsalis, and progressive muscular atrophy.[17] This presentation can also just be weakness causing diminished reflexes.

Is There Any Pain Present? The typical features of muscle disease are weakness, potentially relative to exercise, and, on occasions, muscle pain (myalgia). Conversely, a short history of painful weakness in adulthood, would suggest an inflammatory myositis.[211]

Is The Problem Acute Or Chronic? The age and rate of progression are often helpful in determining the type of muscle disease. Progressive slow weakness absent pain from childhood would suggest degenerative muscular dystrophy. Dreifuss muscular dystrophy and Bethlem myopathy cause fixed contractors that occur early and represent distinctive features of these diseases.[24]

Is Weakness Primarily Distal Or Proximal? The distribution of weakness is also of assistance in defining the likely type of muscle disease, proximal arm, and leg weakness present in limb-girdle muscular dystrophy.[17]

Is The Weakness Localized Or Diffuse? Muscle weakness may be from a myopathy such as dermatomyositis, an inflammatory disease such as rheumatoid arthritis, a neurologic disorder such as Guillain-Barré syndrome, or an infection such as Lyme disease or trichinosis.[17]

Are There Any Focal Neurological Lesions? Myotonic dystrophy, myasthenia gravis, and progressive muscular atrophy may present with partial ptosis.[17]

Prognosis

Morbidity

People with dystrophies having significant heart involvement may nonetheless have almost average life spans, provided that cardiac complications are monitored and aggressively treated.[99] Maintaining ambulation and careful follow-up for evidence of cardiopulmonary complications are essential for long-term care. Although there have been significant advances in the treatment of muscular dystrophy, the disease is still incurable and cannot be prevented. Most patients die before the age of 30 years due to cardiopulmonary failure.[212] Patients can have a better quality of life with proper care.

Mortality 

Duchenne muscular dystrophy can survive around 20 years, but the condition is fatal in 100% of cases.[212] Walker-Warburg syndrome is the most severe of the congenital muscular dystrophies, causing death by one year of age.[213] Patients with Becker dystrophy have a reduced life expectancy, but most survive into the fourth or fifth decade.[213] Children with Duchenne muscular dystrophy usually succumb to other pulmonary or cardiac causes, and death ensues by the late teens. Only 25% live to age 21.[213] Severe cardiac and respiratory problems develop in the late teen years and into the early 20s. Other causes of death include aspiration of food and acute gastric dilation.[213] Life expectancy is normal in patients with facioscapulohumeral dystrophy.[213] 

Complications

Considerations

  • Anesthesia risk
  • Bone demineralization
  • Cardiopulmonary disease
  • Disability

Anesthesia Risk: Patients with muscular dystrophy tend to do poorly after the administration of general anesthetic and will require intensive postoperative observation.[214]

Bone Demineralization: Osteopenia and osteoporosis due to decreased mobilization and immobilization; this can give rise to fractures and scoliosis.[214]

Cardiopulmonary Disease: Frequent follow-up to eliminate secondary complications such as cardiopulmonary disease or cardiac failure due to cardiomyopathy.[214]

Disability: Disability and dependence on walking aids like wheelchairs due to muscular injuries and contractures.[214]

Enhancing Healthcare Team Outcomes

Skills

Adolescents with muscular dystrophy need an interprofessional plan of care, including consideration to heart and breathing difficulties, weight loss/gain, constipation, rehabilitative/developmental difficulties, psychosocial requirements, and neurologic and orthopedic problems.[214] [Level 2]

Strategy

A regulated method for gathering and communicating data helps to support overall performance across programs, sites, and organizations.[64] [Level 2] Therefore, it further enhances the quality of care and decreases disparities in health care delivery through standardization of care. Clinicians and their institutions are enthusiastic in advancing early diagnosis of muscular dystrophy; thus, the strategic indicator for evaluation might be the proportion of patients who receive genetic testing or are offered genetic testing. Clarifying care goals from the beginning allows other team members a framework from which to define their indicators of success more clearly.[64] [Level 2]

Ethics and Responsibilities

Primary care providers may serve as the arbiter of treatment for the various aspects of the disease as well as other problems. Therefore, all other team members now have one centralized point of contact to align with when making decisions on supportive and assistive measures. This point is important because proper implementation and monitoring is a crucial component of management.[64] [Level 2]

Interprofessional Communication 

Another crucial aspect to the management of patients with muscular dystrophy, especially for those with chronic, progressive, and severe forms, relates to patients' need to be informed about research efforts for "their" disease. Because of Internet access and lay support groups, patients today are more familiarized with their disease than ever before. They want to know about current research and relevant ongoing clinical trials, the role of alternative therapies, and other issues and questions.  

All members of the healthcare team must therefore always be ready to provide solid information about a patient's condition by keeping up with fellow team members about what each member's profession can offer in terms of information. This approach facilitates communication that centers around the patient and less around the disease of a given patient and may involve something as simple as providing patients with information on support groups or informative websites. Patients should be encouraged to participate as much as deemed feasible and practical. It is crucial to help patients and their families shift out of a passive mindset and realize that they have a role in their treatment too.[214] [Level 2]

Care Coordination

Effective management will require an interprofessional approach that includes a diverse team of health care professionals.: Strength team members are those aimed at improving strength (e.g., prednisone, stretching). Supportive team members are those who focus on the problems resulting directly from muscle weakness such as respiratory distress. Symptomatic team members are those whose aim is to treat issues that are not directly related to muscle disease but are part of the condition. Examples include cardiac arrhythmias in myotonic dystrophy; and psychological team members whose goal is to improve the patient's mental outlook and provide up to date information about their diseases. An interprofessional team member approach offers a useful conceptual framework and allows for proper delegation of responsibilities.[214] [Level 2]


Details

Updated:

2/6/2023 2:21:07 PM

References


[1]

Allen DG, Whitehead NP, Froehner SC. Absence of Dystrophin Disrupts Skeletal Muscle Signaling: Roles of Ca2+, Reactive Oxygen Species, and Nitric Oxide in the Development of Muscular Dystrophy. Physiological reviews. 2016 Jan:96(1):253-305. doi: 10.1152/physrev.00007.2015. Epub     [PubMed PMID: 26676145]


[2]

Powers SK, Lynch GS, Murphy KT, Reid MB, Zijdewind I. Disease-Induced Skeletal Muscle Atrophy and Fatigue. Medicine and science in sports and exercise. 2016 Nov:48(11):2307-2319     [PubMed PMID: 27128663]


[3]

Díaz-Manera J, Llauger J, Gallardo E, Illa I. Muscle MRI in muscular dystrophies. Acta myologica : myopathies and cardiomyopathies : official journal of the Mediterranean Society of Myology. 2015 Dec:34(2-3):95-108     [PubMed PMID: 27199536]


[4]

Schilling L, Forst R, Forst J, Fujak A. Orthopaedic Disorders in Myotonic Dystrophy Type 1: descriptive clinical study of 21 patients. BMC musculoskeletal disorders. 2013 Dec 1:14():338. doi: 10.1186/1471-2474-14-338. Epub 2013 Dec 1     [PubMed PMID: 24289806]


[5]

Sutherland DH, Olshen R, Cooper L, Wyatt M, Leach J, Mubarak S, Schultz P. The pathomechanics of gait in Duchenne muscular dystrophy. Developmental medicine and child neurology. 1981 Feb:23(1):3-22     [PubMed PMID: 7202868]


[6]

M King W, Kissel JT. Multidisciplinary approach to the management of myopathies. Continuum (Minneapolis, Minn.). 2013 Dec:19(6 Muscle Disease):1650-73. doi: 10.1212/01.CON.0000440664.34051.4d. Epub     [PubMed PMID: 24305452]


[7]

Angelliaume A, Harper L, Lalioui A, Delgove A, Lefèvre Y. Tailor-made management of thoracic scoliosis with cervical hyperextension in muscular dystrophy. European spine journal : official publication of the European Spine Society, the European Spinal Deformity Society, and the European Section of the Cervical Spine Research Society. 2018 Feb:27(2):264-269. doi: 10.1007/s00586-017-5113-8. Epub 2017 Jun 7     [PubMed PMID: 28593385]


[8]

Kang MJ, Yim HB, Hwang HB. Two cases of myotonic dystrophy manifesting various ophthalmic findings with genetic evaluation. Indian journal of ophthalmology. 2016 Jul:64(7):535-7. doi: 10.4103/0301-4738.190157. Epub     [PubMed PMID: 27609169]

Level 3 (low-level) evidence

[9]

Cruz Guzmán Odel R, Chávez García AL, Rodríguez-Cruz M. Muscular dystrophies at different ages: metabolic and endocrine alterations. International journal of endocrinology. 2012:2012():485376. doi: 10.1155/2012/485376. Epub 2012 Jun 3     [PubMed PMID: 22701119]


[10]

Shahrizaila N, Kinnear WJ, Wills AJ. Respiratory involvement in inherited primary muscle conditions. Journal of neurology, neurosurgery, and psychiatry. 2006 Oct:77(10):1108-15     [PubMed PMID: 16980655]


[11]

Cyrulnik SE, Fee RJ, De Vivo DC, Goldstein E, Hinton VJ. Delayed developmental language milestones in children with Duchenne's muscular dystrophy. The Journal of pediatrics. 2007 May:150(5):474-8     [PubMed PMID: 17452219]


[12]

Andrews JG, Wahl RA. Duchenne and Becker muscular dystrophy in adolescents: current perspectives. Adolescent health, medicine and therapeutics. 2018:9():53-63. doi: 10.2147/AHMT.S125739. Epub 2018 Mar 15     [PubMed PMID: 29588625]

Level 3 (low-level) evidence

[13]

Harris SR, Mickelson ECR, Zwicker JG. Diagnosis and management of developmental coordination disorder. CMAJ : Canadian Medical Association journal = journal de l'Association medicale canadienne. 2015 Jun 16:187(9):659-665. doi: 10.1503/cmaj.140994. Epub 2015 May 25     [PubMed PMID: 26009588]


[14]

Bendixen RM, Houtrow A. Parental Reflections on the Diagnostic Process for Duchenne Muscular Dystrophy: A Qualitative Study. Journal of pediatric health care : official publication of National Association of Pediatric Nurse Associates & Practitioners. 2017 May-Jun:31(3):285-292. doi: 10.1016/j.pedhc.2016.09.002. Epub 2016 Oct 12     [PubMed PMID: 27743907]

Level 2 (mid-level) evidence

[15]

Barohn RJ, Dimachkie MM, Jackson CE. A pattern recognition approach to patients with a suspected myopathy. Neurologic clinics. 2014 Aug:32(3):569-93, vii. doi: 10.1016/j.ncl.2014.04.008. Epub     [PubMed PMID: 25037080]


[16]

Sinha R, Sarkar S, Khaitan T, Dutta S. Duchenne muscular dystrophy: Case report and review. Journal of family medicine and primary care. 2017 Jul-Sep:6(3):654-656. doi: 10.4103/2249-4863.222015. Epub     [PubMed PMID: 29417026]

Level 3 (low-level) evidence

[17]

McDonald CM. Clinical approach to the diagnostic evaluation of hereditary and acquired neuromuscular diseases. Physical medicine and rehabilitation clinics of North America. 2012 Aug:23(3):495-563. doi: 10.1016/j.pmr.2012.06.011. Epub     [PubMed PMID: 22938875]


[18]

Yoshioka M, Yorifuji T, Mituyoshi I. Skewed X inactivation in manifesting carriers of Duchenne muscular dystrophy. Clinical genetics. 1998 Feb:53(2):102-7     [PubMed PMID: 9611069]


[19]

Tay SK, Ong HT, Low PS. Transaminitis in Duchenne's muscular dystrophy. Annals of the Academy of Medicine, Singapore. 2000 Nov:29(6):719-22     [PubMed PMID: 11269976]


[20]

Brinkmeyer-Langford C, Kornegay JN. Comparative Genomics of X-linked Muscular Dystrophies: The Golden Retriever Model. Current genomics. 2013 Aug:14(5):330-42. doi: 10.2174/13892029113149990004. Epub     [PubMed PMID: 24403852]

Level 2 (mid-level) evidence

[21]

Jackson M, Marks L, May GHW, Wilson JB. The genetic basis of disease. Essays in biochemistry. 2018 Dec 3:62(5):643-723. doi: 10.1042/EBC20170053. Epub 2018 Dec 2     [PubMed PMID: 30509934]


[22]

Nozoe KT, Akamine RT, Mazzotti DR, Polesel DN, Grossklauss LF, Tufik S, Andersen ML, Moreira GA. Phenotypic contrasts of Duchenne Muscular Dystrophy in women: Two case reports. Sleep science (Sao Paulo, Brazil). 2016 Jul-Sep:9(3):129-133. doi: 10.1016/j.slsci.2016.07.004. Epub 2016 Aug 18     [PubMed PMID: 28123647]

Level 3 (low-level) evidence

[23]

Lo Mauro A, Aliverti A. Physiology of respiratory disturbances in muscular dystrophies. Breathe (Sheffield, England). 2016 Dec:12(4):318-327. doi: 10.1183/20734735.012716. Epub     [PubMed PMID: 28210319]


[24]

Lovering RM, Porter NC, Bloch RJ. The muscular dystrophies: from genes to therapies. Physical therapy. 2005 Dec:85(12):1372-88     [PubMed PMID: 16305275]


[25]

Mendell JR, Lloyd-Puryear M. Report of MDA muscle disease symposium on newborn screening for Duchenne muscular dystrophy. Muscle & nerve. 2013 Jul:48(1):21-6. doi: 10.1002/mus.23810. Epub 2013 May 29     [PubMed PMID: 23716304]


[26]

Juan-Mateu J, Gonzalez-Quereda L, Rodriguez MJ, Baena M, Verdura E, Nascimento A, Ortez C, Baiget M, Gallano P. DMD Mutations in 576 Dystrophinopathy Families: A Step Forward in Genotype-Phenotype Correlations. PloS one. 2015:10(8):e0135189. doi: 10.1371/journal.pone.0135189. Epub 2015 Aug 18     [PubMed PMID: 26284620]


[27]

Le Rumeur E. Dystrophin and the two related genetic diseases, Duchenne and Becker muscular dystrophies. Bosnian journal of basic medical sciences. 2015 Jul 20:15(3):14-20. doi: 10.17305/bjbms.2015.636. Epub 2015 Jul 20     [PubMed PMID: 26295289]


[28]

Desguerre I, Christov C, Mayer M, Zeller R, Becane HM, Bastuji-Garin S, Leturcq F, Chiron C, Chelly J, Gherardi RK. Clinical heterogeneity of duchenne muscular dystrophy (DMD): definition of sub-phenotypes and predictive criteria by long-term follow-up. PloS one. 2009:4(2):e4347. doi: 10.1371/journal.pone.0004347. Epub 2009 Feb 5     [PubMed PMID: 19194511]


[29]

von der Hagen M, Schallner J, Kaindl AM, Koehler K, Mitzscherling P, Abicht A, Grieben U, Korinthenberg R, Kress W, von Moers A, Müller JS, Schara U, Vorgerd M, Walter MC, Müller-Reible C, Hübner C, Lochmüller H, Huebner A. Facing the genetic heterogeneity in neuromuscular disorders: linkage analysis as an economic diagnostic approach towards the molecular diagnosis. Neuromuscular disorders : NMD. 2006 Jan:16(1):4-13     [PubMed PMID: 16378727]


[30]

Grimm T, Meng G, Liechti-Gallati S, Bettecken T, Müller CR, Müller B. On the origin of deletions and point mutations in Duchenne muscular dystrophy: most deletions arise in oogenesis and most point mutations result from events in spermatogenesis. Journal of medical genetics. 1994 Mar:31(3):183-6     [PubMed PMID: 8014964]


[31]

Cooper DN, Krawczak M, Polychronakos C, Tyler-Smith C, Kehrer-Sawatzki H. Where genotype is not predictive of phenotype: towards an understanding of the molecular basis of reduced penetrance in human inherited disease. Human genetics. 2013 Oct:132(10):1077-130. doi: 10.1007/s00439-013-1331-2. Epub 2013 Jul 3     [PubMed PMID: 23820649]

Level 3 (low-level) evidence

[32]

Gao QQ, McNally EM. The Dystrophin Complex: Structure, Function, and Implications for Therapy. Comprehensive Physiology. 2015 Jul 1:5(3):1223-39. doi: 10.1002/cphy.c140048. Epub     [PubMed PMID: 26140716]


[33]

Matsuo M, Awano H, Matsumoto M, Nagai M, Kawaguchi T, Zhang Z, Nishio H. Dystrophin Dp116: A yet to Be Investigated Product of the Duchenne Muscular Dystrophy Gene. Genes. 2017 Oct 2:8(10):. doi: 10.3390/genes8100251. Epub 2017 Oct 2     [PubMed PMID: 28974057]


[34]

Buzin CH, Feng J, Yan J, Scaringe W, Liu Q, den Dunnen J, Mendell JR, Sommer SS. Mutation rates in the dystrophin gene: a hotspot of mutation at a CpG dinucleotide. Human mutation. 2005 Feb:25(2):177-88     [PubMed PMID: 15643612]


[35]

Windpassinger C, Schoser B, Straub V, Hochmeister S, Noor A, Lohberger B, Farra N, Petek E, Schwarzbraun T, Ofner L, Löscher WN, Wagner K, Lochmüller H, Vincent JB, Quasthoff S. An X-linked myopathy with postural muscle atrophy and generalized hypertrophy, termed XMPMA, is caused by mutations in FHL1. American journal of human genetics. 2008 Jan:82(1):88-99. doi: 10.1016/j.ajhg.2007.09.004. Epub     [PubMed PMID: 18179888]


[36]

Raffaele Di Barletta M, Ricci E, Galluzzi G, Tonali P, Mora M, Morandi L, Romorini A, Voit T, Orstavik KH, Merlini L, Trevisan C, Biancalana V, Housmanowa-Petrusewicz I, Bione S, Ricotti R, Schwartz K, Bonne G, Toniolo D. Different mutations in the LMNA gene cause autosomal dominant and autosomal recessive Emery-Dreifuss muscular dystrophy. American journal of human genetics. 2000 Apr:66(4):1407-12     [PubMed PMID: 10739764]


[37]

Mahmood OA, Jiang XM. Limb-girdle muscular dystrophies: where next after six decades from the first proposal (Review). Molecular medicine reports. 2014 May:9(5):1515-32. doi: 10.3892/mmr.2014.2048. Epub 2014 Mar 13     [PubMed PMID: 24626787]


[38]

Mahmood OA, Jiang X, Zhang Q. Limb-girdle muscular dystrophy subtypes: First-reported cohort from northeastern China. Neural regeneration research. 2013 Jul 15:8(20):1907-18. doi: 10.3969/j.issn.1673-5374.2013.20.010. Epub     [PubMed PMID: 25206500]


[39]

Rocha CT, Hoffman EP. Limb-girdle and congenital muscular dystrophies: current diagnostics, management, and emerging technologies. Current neurology and neuroscience reports. 2010 Jul:10(4):267-76. doi: 10.1007/s11910-010-0119-1. Epub     [PubMed PMID: 20467841]


[40]

Salmikangas P, van der Ven PF, Lalowski M, Taivainen A, Zhao F, Suila H, Schröder R, Lappalainen P, Fürst DO, Carpén O. Myotilin, the limb-girdle muscular dystrophy 1A (LGMD1A) protein, cross-links actin filaments and controls sarcomere assembly. Human molecular genetics. 2003 Jan 15:12(2):189-203     [PubMed PMID: 12499399]


[41]

Ki CS, Hong JS, Jeong GY, Ahn KJ, Choi KM, Kim DK, Kim JW. Identification of lamin A/C ( LMNA) gene mutations in Korean patients with autosomal dominant Emery-Dreifuss muscular dystrophy and limb-girdle muscular dystrophy 1B. Journal of human genetics. 2002:47(5):225-8     [PubMed PMID: 12032588]


[42]

Minetti C, Sotgia F, Bruno C, Scartezzini P, Broda P, Bado M, Masetti E, Mazzocco M, Egeo A, Donati MA, Volonte D, Galbiati F, Cordone G, Bricarelli FD, Lisanti MP, Zara F. Mutations in the caveolin-3 gene cause autosomal dominant limb-girdle muscular dystrophy. Nature genetics. 1998 Apr:18(4):365-8     [PubMed PMID: 9537420]


[43]

Peddareddygari LR, Surgan V, Grewal RP. Limb-girdle muscular dystrophy type 2A resulting from homozygous G2338C transversion mutation in the calpain-3 gene. Journal of clinical neuromuscular disease. 2010 Dec:12(2):62-5. doi: 10.1097/CND.0b013e3181f3dbd3. Epub     [PubMed PMID: 21386772]


[44]

Cacciottolo M, Numitone G, Aurino S, Caserta IR, Fanin M, Politano L, Minetti C, Ricci E, Piluso G, Angelini C, Nigro V. Muscular dystrophy with marked Dysferlin deficiency is consistently caused by primary dysferlin gene mutations. European journal of human genetics : EJHG. 2011 Sep:19(9):974-80. doi: 10.1038/ejhg.2011.70. Epub 2011 Apr 27     [PubMed PMID: 21522182]

Level 3 (low-level) evidence

[45]

Okizuka Y, Takeshima Y, Itoh K, Zhang Z, Awano H, Maruyama K, Kumagai T, Yagi M, Matsuo M. Low incidence of limb-girdle muscular dystrophy type 2C revealed by a mutation study in Japanese patients clinically diagnosed with DMD. BMC medical genetics. 2010 Mar 30:11():49. doi: 10.1186/1471-2350-11-49. Epub 2010 Mar 30     [PubMed PMID: 20350330]


[46]

Diniz G, Tosun Yildirim H, Gokben S, Serdaroglu G, Hazan F, Yararbas K, Tukun A. Concomitant alpha- and gamma-sarcoglycan deficiencies in a Turkish boy with a novel deletion in the alpha-sarcoglycan gene. Case reports in genetics. 2014:2014():248561. doi: 10.1155/2014/248561. Epub 2014 Jun 22     [PubMed PMID: 25050186]

Level 3 (low-level) evidence

[47]

Ghafouri-Fard S, Hashemi-Gorji F, Fardaei M, Miryounesi M. Limb Girdle Muscular Dystrophy Type 2E Due to a Novel Large Deletion in SGCB Gene. Iranian journal of child neurology. 2017 Summer:11(3):57-60     [PubMed PMID: 28883879]


[48]

Nigro V, de Sá Moreira E, Piluso G, Vainzof M, Belsito A, Politano L, Puca AA, Passos-Bueno MR, Zatz M. Autosomal recessive limb-girdle muscular dystrophy, LGMD2F, is caused by a mutation in the delta-sarcoglycan gene. Nature genetics. 1996 Oct:14(2):195-8     [PubMed PMID: 8841194]


[49]

Moreira ES, Wiltshire TJ, Faulkner G, Nilforoushan A, Vainzof M, Suzuki OT, Valle G, Reeves R, Zatz M, Passos-Bueno MR, Jenne DE. Limb-girdle muscular dystrophy type 2G is caused by mutations in the gene encoding the sarcomeric protein telethonin. Nature genetics. 2000 Feb:24(2):163-6     [PubMed PMID: 10655062]


[50]

Frosk P, Weiler T, Nylen E, Sudha T, Greenberg CR, Morgan K, Fujiwara TM, Wrogemann K. Limb-girdle muscular dystrophy type 2H associated with mutation in TRIM32, a putative E3-ubiquitin-ligase gene. American journal of human genetics. 2002 Mar:70(3):663-72     [PubMed PMID: 11822024]


[51]

Yamamoto LU, Velloso FJ, Lima BL, Fogaça LL, de Paula F, Vieira NM, Zatz M, Vainzof M. Muscle protein alterations in LGMD2I patients with different mutations in the Fukutin-related protein gene. The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society. 2008 Nov:56(11):995-1001. doi: 10.1369/jhc.2008.951772. Epub 2008 Jul 21     [PubMed PMID: 18645206]


[52]

van der Maarel SM, Deidda G, Lemmers RJ, van Overveld PG, van der Wielen M, Hewitt JE, Sandkuijl L, Bakker B, van Ommen GJ, Padberg GW, Frants RR. De novo facioscapulohumeral muscular dystrophy: frequent somatic mosaicism, sex-dependent phenotype, and the role of mitotic transchromosomal repeat interaction between chromosomes 4 and 10. American journal of human genetics. 2000 Jan:66(1):26-35     [PubMed PMID: 10631134]


[53]

Zernov N, Skoblov M. Genotype-phenotype correlations in FSHD. BMC medical genomics. 2019 Mar 13:12(Suppl 2):43. doi: 10.1186/s12920-019-0488-5. Epub 2019 Mar 13     [PubMed PMID: 30871534]


[54]

Gurzau AD, Chen K, Xue S, Dai W, Lucet IS, Ly TTN, Reversade B, Blewitt ME, Murphy JM. FSHD2- and BAMS-associated mutations confer opposing effects on SMCHD1 function. The Journal of biological chemistry. 2018 Jun 22:293(25):9841-9853. doi: 10.1074/jbc.RA118.003104. Epub 2018 May 10     [PubMed PMID: 29748383]


[55]

Botta A, Vallo L, Rinaldi F, Bonifazi E, Amati F, Biancolella M, Gambardella S, Mancinelli E, Angelini C, Meola G, Novelli G. Gene expression analysis in myotonic dystrophy: indications for a common molecular pathogenic pathway in DM1 and DM2. Gene expression. 2007:13(6):339-51     [PubMed PMID: 17708420]


[56]

Llamusí B, Artero R. Molecular Effects of the CTG Repeats in Mutant Dystrophia Myotonica Protein Kinase Gene. Current genomics. 2008 Dec:9(8):509-16. doi: 10.2174/138920208786847944. Epub     [PubMed PMID: 19516957]


[57]

Yum K, Wang ET, Kalsotra A. Myotonic dystrophy: disease repeat range, penetrance, age of onset, and relationship between repeat size and phenotypes. Current opinion in genetics & development. 2017 Jun:44():30-37. doi: 10.1016/j.gde.2017.01.007. Epub 2017 Feb 14     [PubMed PMID: 28213156]

Level 3 (low-level) evidence

[58]

Paulson H. Repeat expansion diseases. Handbook of clinical neurology. 2018:147():105-123. doi: 10.1016/B978-0-444-63233-3.00009-9. Epub     [PubMed PMID: 29325606]


[59]

Ricker K, Grimm T, Koch MC, Schneider C, Kress W, Reimers CD, Schulte-Mattler W, Mueller-Myhsok B, Toyka KV, Mueller CR. Linkage of proximal myotonic myopathy to chromosome 3q. Neurology. 1999 Jan 1:52(1):170-1     [PubMed PMID: 9921867]


[60]

Brais B, Rouleau GA, Bouchard JP, Fardeau M, Tomé FM. Oculopharyngeal muscular dystrophy. Seminars in neurology. 1999:19(1):59-66     [PubMed PMID: 10711989]


[61]

Rahimov F, Kunkel LM. The cell biology of disease: cellular and molecular mechanisms underlying muscular dystrophy. The Journal of cell biology. 2013 May 13:201(4):499-510. doi: 10.1083/jcb.201212142. Epub     [PubMed PMID: 23671309]


[62]

Bellayou H, Hamzi K, Rafai MA, Karkouri M, Slassi I, Azeddoug H, Nadifi S. Duchenne and Becker muscular dystrophy: contribution of a molecular and immunohistochemical analysis in diagnosis in Morocco. Journal of biomedicine & biotechnology. 2009:2009():325210. doi: 10.1155/2009/325210. Epub 2009 May 19     [PubMed PMID: 19461958]


[63]

Aartsma-Rus A, Van Deutekom JC, Fokkema IF, Van Ommen GJ, Den Dunnen JT. Entries in the Leiden Duchenne muscular dystrophy mutation database: an overview of mutation types and paradoxical cases that confirm the reading-frame rule. Muscle & nerve. 2006 Aug:34(2):135-44     [PubMed PMID: 16770791]

Level 3 (low-level) evidence

[64]

Mah JK. Current and emerging treatment strategies for Duchenne muscular dystrophy. Neuropsychiatric disease and treatment. 2016:12():1795-807. doi: 10.2147/NDT.S93873. Epub 2016 Jul 22     [PubMed PMID: 27524897]


[65]

Jacobs PA, Hunt PA, Mayer M, Bart RD. Duchenne muscular dystrophy (DMD) in a female with an X/autosome translocation: further evidence that the DMD locus is at Xp21. American journal of human genetics. 1981 Jul:33(4):513-8     [PubMed PMID: 7258185]


[66]

Theadom A, Rodrigues M, Roxburgh R, Balalla S, Higgins C, Bhattacharjee R, Jones K, Krishnamurthi R, Feigin V. Prevalence of muscular dystrophies: a systematic literature review. Neuroepidemiology. 2014:43(3-4):259-68. doi: 10.1159/000369343. Epub 2014 Dec 16     [PubMed PMID: 25532075]

Level 1 (high-level) evidence

[67]

Mah JK, Korngut L, Fiest KM, Dykeman J, Day LJ, Pringsheim T, Jette N. A Systematic Review and Meta-analysis on the Epidemiology of the Muscular Dystrophies. The Canadian journal of neurological sciences. Le journal canadien des sciences neurologiques. 2016 Jan:43(1):163-77. doi: 10.1017/cjn.2015.311. Epub     [PubMed PMID: 26786644]

Level 1 (high-level) evidence

[68]

Suominen T, Bachinski LL, Auvinen S, Hackman P, Baggerly KA, Angelini C, Peltonen L, Krahe R, Udd B. Population frequency of myotonic dystrophy: higher than expected frequency of myotonic dystrophy type 2 (DM2) mutation in Finland. European journal of human genetics : EJHG. 2011 Jul:19(7):776-82. doi: 10.1038/ejhg.2011.23. Epub 2011 Mar 2     [PubMed PMID: 21364698]


[69]

Ashizawa T, Gagnon C, Groh WJ, Gutmann L, Johnson NE, Meola G, Moxley R 3rd, Pandya S, Rogers MT, Simpson E, Angeard N, Bassez G, Berggren KN, Bhakta D, Bozzali M, Broderick A, Byrne JLB, Campbell C, Cup E, Day JW, De Mattia E, Duboc D, Duong T, Eichinger K, Ekstrom AB, van Engelen B, Esparis B, Eymard B, Ferschl M, Gadalla SM, Gallais B, Goodglick T, Heatwole C, Hilbert J, Holland V, Kierkegaard M, Koopman WJ, Lane K, Maas D, Mankodi A, Mathews KD, Monckton DG, Moser D, Nazarian S, Nguyen L, Nopoulos P, Petty R, Phetteplace J, Puymirat J, Raman S, Richer L, Roma E, Sampson J, Sansone V, Schoser B, Sterling L, Statland J, Subramony SH, Tian C, Trujillo C, Tomaselli G, Turner C, Venance S, Verma A, White M, Winblad S. Consensus-based care recommendations for adults with myotonic dystrophy type 1. Neurology. Clinical practice. 2018 Dec:8(6):507-520. doi: 10.1212/CPJ.0000000000000531. Epub     [PubMed PMID: 30588381]

Level 3 (low-level) evidence

[70]

Stark AE. Determinants of the incidence of Duchenne muscular dystrophy. Annals of translational medicine. 2015 Nov:3(19):287. doi: 10.3978/j.issn.2305-5839.2015.10.45. Epub     [PubMed PMID: 26697447]


[71]

Mah JK, Korngut L, Dykeman J, Day L, Pringsheim T, Jette N. A systematic review and meta-analysis on the epidemiology of Duchenne and Becker muscular dystrophy. Neuromuscular disorders : NMD. 2014 Jun:24(6):482-91. doi: 10.1016/j.nmd.2014.03.008. Epub 2014 Mar 22     [PubMed PMID: 24780148]

Level 1 (high-level) evidence

[72]

Bushby KM, Thambyayah M, Gardner-Medwin D. Prevalence and incidence of Becker muscular dystrophy. Lancet (London, England). 1991 Apr 27:337(8748):1022-4     [PubMed PMID: 1673177]


[73]

Mostacciuolo ML, Miorin M, Martinello F, Angelini C, Perini P, Trevisan CP. Genetic epidemiology of congenital muscular dystrophy in a sample from north-east Italy. Human genetics. 1996 Mar:97(3):277-9     [PubMed PMID: 8786062]


[74]

Graziano A, Bianco F, D'Amico A, Moroni I, Messina S, Bruno C, Pegoraro E, Mora M, Astrea G, Magri F, Comi GP, Berardinelli A, Moggio M, Morandi L, Pini A, Petillo R, Tasca G, Monforte M, Minetti C, Mongini T, Ricci E, Gorni K, Battini R, Villanova M, Politano L, Gualandi F, Ferlini A, Muntoni F, Santorelli FM, Bertini E, Pane M, Mercuri E. Prevalence of congenital muscular dystrophy in Italy: a population study. Neurology. 2015 Mar 3:84(9):904-11. doi: 10.1212/WNL.0000000000001303. Epub 2015 Feb 4     [PubMed PMID: 25653289]


[75]

Peterlin B, Zidar J, Meznaric-Petrusa M, Zupancic N. Genetic epidemiology of Duchenne and Becker muscular dystrophy in Slovenia. Clinical genetics. 1997 Feb:51(2):94-7     [PubMed PMID: 9111995]


[76]

Norwood FL, Harling C, Chinnery PF, Eagle M, Bushby K, Straub V. Prevalence of genetic muscle disease in Northern England: in-depth analysis of a muscle clinic population. Brain : a journal of neurology. 2009 Nov:132(Pt 11):3175-86. doi: 10.1093/brain/awp236. Epub 2009 Sep 18     [PubMed PMID: 19767415]


[77]

Jacobson C, Côté PD, Rossi SG, Rotundo RL, Carbonetto S. The dystroglycan complex is necessary for stabilization of acetylcholine receptor clusters at neuromuscular junctions and formation of the synaptic basement membrane. The Journal of cell biology. 2001 Feb 5:152(3):435-50     [PubMed PMID: 11157973]


[78]

Montanaro F, Lindenbaum M, Carbonetto S. alpha-Dystroglycan is a laminin receptor involved in extracellular matrix assembly on myotubes and muscle cell viability. The Journal of cell biology. 1999 Jun 14:145(6):1325-40     [PubMed PMID: 10366602]


[79]

Shivers RR, Atkinson BG. The dystrophic murine skeletal muscle cell plasma membrane is structurally intact but "leaky" to creatine phosphokinase. A freeze-fracture analysis. The American journal of pathology. 1984 Sep:116(3):482-96     [PubMed PMID: 6476081]


[80]

Davis MH, Cappel R, Vester JW, Samaha FJ, Gruenstein E. Creatine kinase activity in normal and Duchenne muscular dystrophy fibroblasts. Muscle & nerve. 1982 Jan:5(1):1-6     [PubMed PMID: 7057800]


[81]

Kuller JA, Hoffman EP, Fries MH, Golbus MS. Prenatal diagnosis of Duchenne muscular dystrophy by fetal muscle biopsy. Human genetics. 1992 Sep-Oct:90(1-2):34-40     [PubMed PMID: 1427785]


[82]

Adams JC, Brancaccio A. The evolution of the dystroglycan complex, a major mediator of muscle integrity. Biology open. 2015 Aug 28:4(9):1163-79. doi: 10.1242/bio.012468. Epub 2015 Aug 28     [PubMed PMID: 26319583]


[83]

Barresi R, Campbell KP. Dystroglycan: from biosynthesis to pathogenesis of human disease. Journal of cell science. 2006 Jan 15:119(Pt 2):199-207     [PubMed PMID: 16410545]


[84]

Kharraz Y, Guerra J, Pessina P, Serrano AL, Muñoz-Cánoves P. Understanding the process of fibrosis in Duchenne muscular dystrophy. BioMed research international. 2014:2014():965631. doi: 10.1155/2014/965631. Epub 2014 May 4     [PubMed PMID: 24877152]

Level 3 (low-level) evidence

[85]

Klingler W, Jurkat-Rott K, Lehmann-Horn F, Schleip R. The role of fibrosis in Duchenne muscular dystrophy. Acta myologica : myopathies and cardiomyopathies : official journal of the Mediterranean Society of Myology. 2012 Dec:31(3):184-95     [PubMed PMID: 23620650]


[86]

Peric S, Nisic T, Milicev M, Basta I, Marjanovic I, Peric M, Lavrnic D, Rakocevic Stojanovic V. Hypogonadism and erectile dysfunction in myotonic dystrophy type 1. Acta myologica : myopathies and cardiomyopathies : official journal of the Mediterranean Society of Myology. 2013 Oct:32(2):106-9     [PubMed PMID: 24399868]


[87]

Toussaint M, Davidson Z, Bouvoie V, Evenepoel N, Haan J, Soudon P. Dysphagia in Duchenne muscular dystrophy: practical recommendations to guide management. Disability and rehabilitation. 2016 Oct:38(20):2052-62. doi: 10.3109/09638288.2015.1111434. Epub 2016 Jan 5     [PubMed PMID: 26728920]


[88]

Nereo NE, Fee RJ, Hinton VJ. Parental stress in mothers of boys with duchenne muscular dystrophy. Journal of pediatric psychology. 2003 Oct-Nov:28(7):473-84     [PubMed PMID: 12968039]


[89]

Birnkrant DJ, Bushby K, Bann CM, Apkon SD, Blackwell A, Brumbaugh D, Case LE, Clemens PR, Hadjiyannakis S, Pandya S, Street N, Tomezsko J, Wagner KR, Ward LM, Weber DR, DMD Care Considerations Working Group. Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and neuromuscular, rehabilitation, endocrine, and gastrointestinal and nutritional management. The Lancet. Neurology. 2018 Mar:17(3):251-267. doi: 10.1016/S1474-4422(18)30024-3. Epub 2018 Feb 3     [PubMed PMID: 29395989]

Level 3 (low-level) evidence

[90]

Ali SZ, Taguchi A, Rosenberg H. Malignant hyperthermia. Best practice & research. Clinical anaesthesiology. 2003 Dec:17(4):519-33     [PubMed PMID: 14661655]


[91]

Rosenberg H, Davis M, James D, Pollock N, Stowell K. Malignant hyperthermia. Orphanet journal of rare diseases. 2007 Apr 24:2():21     [PubMed PMID: 17456235]


[92]

Takami Y, Takeshima Y, Awano H, Okizuka Y, Yagi M, Matsuo M. High incidence of electrocardiogram abnormalities in young patients with duchenne muscular dystrophy. Pediatric neurology. 2008 Dec:39(6):399-403. doi: 10.1016/j.pediatrneurol.2008.08.006. Epub     [PubMed PMID: 19027585]


[93]

Adam MP, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, Bird TD. Myotonic Dystrophy Type 1. GeneReviews(®). 1993:():     [PubMed PMID: 20301344]


[94]

Pelargonio G, Dello Russo A, Sanna T, De Martino G, Bellocci F. Myotonic dystrophy and the heart. Heart (British Cardiac Society). 2002 Dec:88(6):665-70     [PubMed PMID: 12433913]


[95]

Rajdev A, Groh WJ. Arrhythmias in the muscular dystrophies. Cardiac electrophysiology clinics. 2015 Jun:7(2):303-8. doi: 10.1016/j.ccep.2015.03.011. Epub 2015 Mar 29     [PubMed PMID: 26002394]


[96]

Wollinsky KH, Kutter B, Geiger PM. Long-term ventilation of patients with Duchenne muscular dystrophy: experiences at the Neuromuscular Centre Ulm. Acta myologica : myopathies and cardiomyopathies : official journal of the Mediterranean Society of Myology. 2012 Dec:31(3):170-8     [PubMed PMID: 23620648]


[97]

Mathew T, Williams L, Navaratnam G, Rana B, Wheeler R, Collins K, Harkness A, Jones R, Knight D, O'Gallagher K, Oxborough D, Ring L, Sandoval J, Stout M, Sharma V, Steeds RP, British Society of Echocardiography Education Committee. Diagnosis and assessment of dilated cardiomyopathy: a guideline protocol from the British Society of Echocardiography. Echo research and practice. 2017 Jun:4(2):G1-G13. doi: 10.1530/ERP-16-0037. Epub     [PubMed PMID: 28592613]


[98]

Mestroni L, Brun F, Spezzacatene A, Sinagra G, Taylor MR. GENETIC CAUSES OF DILATED CARDIOMYOPATHY. Progress in pediatric cardiology. 2014 Dec:37(1-2):13-18     [PubMed PMID: 25584016]


[99]

Verhaert D, Richards K, Rafael-Fortney JA, Raman SV. Cardiac involvement in patients with muscular dystrophies: magnetic resonance imaging phenotype and genotypic considerations. Circulation. Cardiovascular imaging. 2011 Jan:4(1):67-76. doi: 10.1161/CIRCIMAGING.110.960740. Epub     [PubMed PMID: 21245364]


[100]

Mavrogeni SI, Markousis-Mavrogenis G, Papavasiliou A, Papadopoulos G, Kolovou G. Cardiac Involvement in Duchenne Muscular Dystrophy and Related Dystrophinopathies. Methods in molecular biology (Clifton, N.J.). 2018:1687():31-42. doi: 10.1007/978-1-4939-7374-3_3. Epub     [PubMed PMID: 29067654]


[101]

Oldfors A, Eriksson BO, Kyllerman M, Martinsson T, Wahlström J. Dilated cardiomyopathy and the dystrophin gene: an illustrated review. British heart journal. 1994 Oct:72(4):344-8     [PubMed PMID: 7833192]


[102]

Connuck DM, Sleeper LA, Colan SD, Cox GF, Towbin JA, Lowe AM, Wilkinson JD, Orav EJ, Cuniberti L, Salbert BA, Lipshultz SE, Pediatric Cardiomyopathy Registry Study Group. Characteristics and outcomes of cardiomyopathy in children with Duchenne or Becker muscular dystrophy: a comparative study from the Pediatric Cardiomyopathy Registry. American heart journal. 2008 Jun:155(6):998-1005. doi: 10.1016/j.ahj.2008.01.018. Epub 2008 Mar 19     [PubMed PMID: 18513510]

Level 2 (mid-level) evidence

[103]

Skalsky AJ, McDonald CM. Prevention and management of limb contractures in neuromuscular diseases. Physical medicine and rehabilitation clinics of North America. 2012 Aug:23(3):675-87. doi: 10.1016/j.pmr.2012.06.009. Epub     [PubMed PMID: 22938881]


[104]

Choi YA, Chun SM, Kim Y, Shin HI. Lower extremity joint contracture according to ambulatory status in children with Duchenne muscular dystrophy. BMC musculoskeletal disorders. 2018 Aug 16:19(1):287. doi: 10.1186/s12891-018-2212-6. Epub 2018 Aug 16     [PubMed PMID: 30111310]


[105]

Ricci G, Zatz M, Tupler R. Facioscapulohumeral Muscular Dystrophy: More Complex than it Appears. Current molecular medicine. 2014:14(8):1052-1068. doi: 10.2174/1566524014666141010155054. Epub     [PubMed PMID: 25323867]


[106]

Sackley C, Disler PB, Turner-Stokes L, Wade DT, Brittle N, Hoppitt T. Rehabilitation interventions for foot drop in neuromuscular disease. The Cochrane database of systematic reviews. 2009 Jul 8:(3):CD003908. doi: 10.1002/14651858.CD003908.pub3. Epub 2009 Jul 8     [PubMed PMID: 19588347]

Level 1 (high-level) evidence

[107]

Gissy JJ, Johnson T, Fox DJ, Kumar A, Ciafaloni E, van Essen AJ, Peay HL, Martin A, Lucas A, Finkel RS, MD STARnet. Delayed onset of ambulation in boys with Duchenne muscular dystrophy: Potential use as an endpoint in clinical trials. Neuromuscular disorders : NMD. 2017 Oct:27(10):905-910. doi: 10.1016/j.nmd.2017.06.002. Epub 2017 Jul 21     [PubMed PMID: 28739181]


[108]

Hahn C, Salajegheh MK. Myotonic disorders: A review article. Iranian journal of neurology. 2016 Jan 5:15(1):46-53     [PubMed PMID: 27141276]


[109]

Barzegar M, Niknam E, Habibi P, Shiva S, Tahmasebi S. Bone Mineral Density and Bone Metabolism in Patients with Duchenne Muscular Dystrophy. Iranian journal of child neurology. 2018 Winter:12(1):77-83     [PubMed PMID: 29379565]


[110]

Partridge T. Could exon skipping help dystrophic boys to run, hop, and jump? Molecular therapy : the journal of the American Society of Gene Therapy. 2014 Nov:22(11):1884-6. doi: 10.1038/mt.2014.189. Epub     [PubMed PMID: 25365985]


[111]

Chang RF, Mubarak SJ. Pathomechanics of Gowers' sign: a video analysis of a spectrum of Gowers' maneuvers. Clinical orthopaedics and related research. 2012 Jul:470(7):1987-91. doi: 10.1007/s11999-011-2210-6. Epub 2011 Dec 28     [PubMed PMID: 22203329]


[112]

Kornegay JN, Childers MK, Bogan DJ, Bogan JR, Nghiem P, Wang J, Fan Z, Howard JF Jr, Schatzberg SJ, Dow JL, Grange RW, Styner MA, Hoffman EP, Wagner KR. The paradox of muscle hypertrophy in muscular dystrophy. Physical medicine and rehabilitation clinics of North America. 2012 Feb:23(1):149-72, xii. doi: 10.1016/j.pmr.2011.11.014. Epub     [PubMed PMID: 22239881]


[113]

Koenig M, Beggs AH, Moyer M, Scherpf S, Heindrich K, Bettecken T, Meng G, Müller CR, Lindlöf M, Kaariainen H, de la Chapellet A, Kiuru A, Savontaus ML, Gilgenkrantz H, Récan D, Chelly J, Kaplan JC, Covone AE, Archidiacono N, Romeo G, Liechti-Gailati S, Schneider V, Braga S, Moser H, Darras BT, Murphy P, Francke U, Chen JD, Morgan G, Denton M, Greenberg CR, Wrogemann K, Blonden LA, van Paassen MB, van Ommen GJ, Kunkel LM. The molecular basis for Duchenne versus Becker muscular dystrophy: correlation of severity with type of deletion. American journal of human genetics. 1989 Oct:45(4):498-506     [PubMed PMID: 2491009]


[114]

Mateos-Aierdi AJ, Goicoechea M, Aiastui A, Fernández-Torrón R, Garcia-Puga M, Matheu A, López de Munain A. Muscle wasting in myotonic dystrophies: a model of premature aging. Frontiers in aging neuroscience. 2015:7():125. doi: 10.3389/fnagi.2015.00125. Epub 2015 Jul 9     [PubMed PMID: 26217220]


[115]

Reimers CD, Schlotter B, Eicke BM, Witt TN. Calf enlargement in neuromuscular diseases: a quantitative ultrasound study in 350 patients and review of the literature. Journal of the neurological sciences. 1996 Nov:143(1-2):46-56     [PubMed PMID: 8981297]


[116]

Marden FA, Connolly AM, Siegel MJ, Rubin DA. Compositional analysis of muscle in boys with Duchenne muscular dystrophy using MR imaging. Skeletal radiology. 2005 Mar:34(3):140-8     [PubMed PMID: 15538561]


[117]

Villanova M, Mercuri E, Bertini E, Sabatelli P, Morandi L, Mora M, Sewry C, Brockington M, Brown SC, Ferreiro A, Maraldi NM, Toda T, Guicheney P, Merlini L, Muntoni F. Congenital muscular dystrophy associated with calf hypertrophy, microcephaly and severe mental retardation in three Italian families: evidence for a novel CMD syndrome. Neuromuscular disorders : NMD. 2000 Dec:10(8):541-7     [PubMed PMID: 11053679]


[118]

Lacomis D. Electrodiagnostic approach to the patient with suspected myopathy. Neurologic clinics. 2002 May:20(2):587-603     [PubMed PMID: 12152448]


[119]

Adam MP, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, Pegoraro E, Hoffman EP. Limb-Girdle Muscular Dystrophy Overview – RETIRED CHAPTER, FOR HISTORICAL REFERENCE ONLY. GeneReviews(®). 1993:():     [PubMed PMID: 20301582]

Level 3 (low-level) evidence

[120]

Forst R, Forst J, Heller KD, Hengstler K. [Characteristics in the treatment of scoliosis in muscular diseases]. Zeitschrift fur Orthopadie und ihre Grenzgebiete. 1997 Mar-Apr:135(2):95-105     [PubMed PMID: 9214180]


[121]

Ropars J, Lempereur M, Vuillerot C, Tiffreau V, Peudenier S, Cuisset JM, Pereon Y, Leboeuf F, Delporte L, Delpierre Y, Gross R, Brochard S. Muscle Activation during Gait in Children with Duchenne Muscular Dystrophy. PloS one. 2016:11(9):e0161938. doi: 10.1371/journal.pone.0161938. Epub 2016 Sep 13     [PubMed PMID: 27622734]


[122]

Gijsbertse K, Goselink R, Lassche S, Nillesen M, Sprengers A, Verdonschot N, van Alfen N, de Korte C. Ultrasound Imaging of Muscle Contraction of the Tibialis Anterior in Patients with Facioscapulohumeral Dystrophy. Ultrasound in medicine & biology. 2017 Nov:43(11):2537-2545. doi: 10.1016/j.ultrasmedbio.2017.06.016. Epub 2017 Jul 29     [PubMed PMID: 28764967]


[123]

Angelini C, Pinzan E. Advances in imaging of brain abnormalities in neuromuscular disease. Therapeutic advances in neurological disorders. 2019:12():1756286419845567. doi: 10.1177/1756286419845567. Epub 2019 May 6     [PubMed PMID: 31105770]

Level 3 (low-level) evidence

[124]

D'Angelo MG, Lorusso ML, Civati F, Comi GP, Magri F, Del Bo R, Guglieri M, Molteni M, Turconi AC, Bresolin N. Neurocognitive profiles in Duchenne muscular dystrophy and gene mutation site. Pediatric neurology. 2011 Nov:45(5):292-9. doi: 10.1016/j.pediatrneurol.2011.08.003. Epub     [PubMed PMID: 22000308]


[125]

Ho G, Cardamone M, Farrar M. Congenital and childhood myotonic dystrophy: Current aspects of disease and future directions. World journal of clinical pediatrics. 2015 Nov 8:4(4):66-80. doi: 10.5409/wjcp.v4.i4.66. Epub 2015 Nov 8     [PubMed PMID: 26566479]

Level 3 (low-level) evidence

[126]

Chamova T, Guergueltcheva V, Raycheva M, Todorov T, Genova J, Bichev S, Bojinova V, Mitev V, Tournev I, Todorova A. Association between loss of dp140 and cognitive impairment in duchenne and becker dystrophies. Balkan journal of medical genetics : BJMG. 2013 Jun:16(1):21-30. doi: 10.2478/bjmg-2013-0014. Epub     [PubMed PMID: 24265581]


[127]

Kumagai T, Miura K, Ohki T, Matsumoto A, Miyazaki S, Nakamura M, Ochi N, Takahashi O. [Central nervous system involvements in Duchenne/Becker muscular dystrophy]. No to hattatsu = Brain and development. 2001 Nov:33(6):480-6     [PubMed PMID: 11725514]


[128]

Gadoth N, Oksenberg A. Sleep and sleep disorders in rare hereditary diseases: a reminder for the pediatrician, pediatric and adult neurologist, general practitioner, and sleep specialist. Frontiers in neurology. 2014:5():133. doi: 10.3389/fneur.2014.00133. Epub 2014 Jul 17     [PubMed PMID: 25101051]


[129]

Echenne B, Rivier F, Jellali AJ, Azais M, Mornet D, Pons F. Merosin positive congenital muscular dystrophy with mental deficiency, epilepsy and MRI changes in the cerebral white matter. Neuromuscular disorders : NMD. 1997 May:7(3):187-90     [PubMed PMID: 9185183]


[130]

Adam MP, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, Sparks SE, Quijano-Roy S, Harper A, Rutkowski A, Gordon E, Hoffman EP, Pegoraro E. Congenital Muscular Dystrophy Overview – RETIRED CHAPTER, FOR HISTORICAL REFERENCE ONLY. GeneReviews(®). 1993:():     [PubMed PMID: 20301468]

Level 3 (low-level) evidence

[131]

Louprasong AC, Light DJ, Diller RS. Spider dystrophy as an ocular manifestation of myotonic dystrophy. Optometry (St. Louis, Mo.). 2010 Apr:81(4):188-93. doi: 10.1016/j.optm.2009.08.013. Epub     [PubMed PMID: 20346890]


[132]

MacLeod M, Kelly R, Robb SA, Borzyskowski M. Bladder dysfunction in Duchenne muscular dystrophy. Archives of disease in childhood. 2003 Apr:88(4):347-9     [PubMed PMID: 12651768]


[133]

Meola G. Clinical aspects, molecular pathomechanisms and management of myotonic dystrophies. Acta myologica : myopathies and cardiomyopathies : official journal of the Mediterranean Society of Myology. 2013 Dec:32(3):154-65     [PubMed PMID: 24803843]


[134]

Reiter C, Gramer E. [Anticipation in patients with iridescent multicoloured posterior capsular lens opacities ("Christmas tree cataract") : The Role in the diagnosis of myotonic dystrophy]. Der Ophthalmologe : Zeitschrift der Deutschen Ophthalmologischen Gesellschaft. 2009 Dec:106(12):1116-20. doi: 10.1007/s00347-009-1924-2. Epub     [PubMed PMID: 19326122]


[135]

Lo Cascio CM, Goetze O, Latshang TD, Bluemel S, Frauenfelder T, Bloch KE. Gastrointestinal Dysfunction in Patients with Duchenne Muscular Dystrophy. PloS one. 2016:11(10):e0163779. doi: 10.1371/journal.pone.0163779. Epub 2016 Oct 13     [PubMed PMID: 27736891]


[136]

Bellini M, Biagi S, Stasi C, Costa F, Mumolo MG, Ricchiuti A, Marchi S. Gastrointestinal manifestations in myotonic muscular dystrophy. World journal of gastroenterology. 2006 Mar 28:12(12):1821-8     [PubMed PMID: 16609987]


[137]

Tack J, Müller-Lissner S, Stanghellini V, Boeckxstaens G, Kamm MA, Simren M, Galmiche JP, Fried M. Diagnosis and treatment of chronic constipation--a European perspective. Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society. 2011 Aug:23(8):697-710. doi: 10.1111/j.1365-2982.2011.01709.x. Epub 2011 May 24     [PubMed PMID: 21605282]

Level 3 (low-level) evidence

[138]

Clavé P, Shaker R. Dysphagia: current reality and scope of the problem. Nature reviews. Gastroenterology & hepatology. 2015 May:12(5):259-70. doi: 10.1038/nrgastro.2015.49. Epub 2015 Apr 7     [PubMed PMID: 25850008]


[139]

Erdenen F, Toros AB, Uzum AK, Sacak S. Steinert's syndrome presenting as anal incontinence: a case report. Journal of medical case reports. 2011 Aug 12:5():371. doi: 10.1186/1752-1947-5-371. Epub 2011 Aug 12     [PubMed PMID: 21838873]

Level 3 (low-level) evidence

[140]

Kim WB, Jeong JY, Doo SW, Yang WJ, Song YS, Lee SR, Park JW, Kim DW. Myotonic dystrophy type 1 presenting as male infertility. Korean journal of urology. 2012 Feb:53(2):134-6. doi: 10.4111/kju.2012.53.2.134. Epub 2012 Feb 20     [PubMed PMID: 22379595]


[141]

Rodríguez-Cruz M, Atilano-Miguel S, Barbosa-Cortés L, Bernabé-García M, Almeida-Becerril T, Cárdenas-Conejo A, Del Rocío Cruz-Guzmán O, Maldonado-Hernández J. Evidence of muscle loss delay and improvement of hyperinsulinemia and insulin resistance in Duchenne muscular dystrophy supplemented with omega-3 fatty acids: A randomized study. Clinical nutrition (Edinburgh, Scotland). 2019 Oct:38(5):2087-2097. doi: 10.1016/j.clnu.2018.10.017. Epub 2018 Oct 30     [PubMed PMID: 30420291]

Level 1 (high-level) evidence

[142]

Dreimann M, Hoffmann M, Kossow K, Hitzl W, Meier O, Koller H. Scoliosis and chest cage deformity measures predicting impairments in pulmonary function: a cross-sectional study of 492 patients with scoliosis to improve the early identification of patients at risk. Spine. 2014 Nov 15:39(24):2024-33. doi: 10.1097/BRS.0000000000000601. Epub     [PubMed PMID: 25202929]

Level 2 (mid-level) evidence

[143]

Wang CH, Bonnemann CG, Rutkowski A, Sejersen T, Bellini J, Battista V, Florence JM, Schara U, Schuler PM, Wahbi K, Aloysius A, Bash RO, Béroud C, Bertini E, Bushby K, Cohn RD, Connolly AM, Deconinck N, Desguerre I, Eagle M, Estournet-Mathiaud B, Ferreiro A, Fujak A, Goemans N, Iannaccone ST, Jouinot P, Main M, Melacini P, Mueller-Felber W, Muntoni F, Nelson LL, Rahbek J, Quijano-Roy S, Sewry C, Storhaug K, Simonds A, Tseng B, Vajsar J, Vianello A, Zeller R, International Standard of Care Committee for Congenital Muscular Dystrophy. Consensus statement on standard of care for congenital muscular dystrophies. Journal of child neurology. 2010 Dec:25(12):1559-81. doi: 10.1177/0883073810381924. Epub 2010 Nov 15     [PubMed PMID: 21078917]

Level 3 (low-level) evidence

[144]

Kinane TB, Mayer OH, Duda PW, Lowes LP, Moody SL, Mendell JR. Long-Term Pulmonary Function in Duchenne Muscular Dystrophy: Comparison of Eteplirsen-Treated Patients to Natural History. Journal of neuromuscular diseases. 2018:5(1):47-58. doi: 10.3233/JND-170272. Epub     [PubMed PMID: 29278896]


[145]

Mangera Z, Panesar G, Makker H. Practical approach to management of respiratory complications in neurological disorders. International journal of general medicine. 2012:5():255-63. doi: 10.2147/IJGM.S26333. Epub 2012 Mar 21     [PubMed PMID: 22505823]


[146]

Howard RS, Davidson C. Long term ventilation in neurogenic respiratory failure. Journal of neurology, neurosurgery, and psychiatry. 2003 Sep:74 Suppl 3(Suppl 3):iii24-30     [PubMed PMID: 12933911]


[147]

Polat M, Sakinci O, Ersoy B, Sezer RG, Yilmaz H. Assessment of sleep-related breathing disorders in patients with duchenne muscular dystrophy. Journal of clinical medicine research. 2012 Oct:4(5):332-7     [PubMed PMID: 23024736]


[148]

Hathout Y, Seol H, Han MH, Zhang A, Brown KJ, Hoffman EP. Clinical utility of serum biomarkers in Duchenne muscular dystrophy. Clinical proteomics. 2016:13():9. doi: 10.1186/s12014-016-9109-x. Epub 2016 Apr 5     [PubMed PMID: 27051355]


[149]

Takasugi T, Ishihara T, Kawamura J, Sasaki K, Toyoda T, Oosumi M, Suzuki K, Nishio K, Aoyagi T, Kawashiro T. [Blood gas changes in Duchenne type muscular dystrophy]. Nihon Kyobu Shikkan Gakkai zasshi. 1995 Jan:33(1):17-22     [PubMed PMID: 7699962]


[150]

Rubegni A, Malandrini A, Dosi C, Astrea G, Baldacci J, Battisti C, Bertocci G, Donati MA, Dotti MT, Federico A, Giannini F, Grosso S, Guerrini R, Lenzi S, Maioli MA, Melani F, Mercuri E, Sacchini M, Salvatore S, Siciliano G, Tolomeo D, Tonin P, Volpi N, Santorelli FM, Cassandrini D. Next-generation sequencing approach to hyperCKemia: A 2-year cohort study. Neurology. Genetics. 2019 Oct:5(5):e352. doi: 10.1212/NXG.0000000000000352. Epub 2019 Aug 16     [PubMed PMID: 31517061]


[151]

Moghadam-Kia S, Oddis CV, Aggarwal R. Approach to asymptomatic creatine kinase elevation. Cleveland Clinic journal of medicine. 2016 Jan:83(1):37-42. doi: 10.3949/ccjm.83a.14120. Epub     [PubMed PMID: 26760521]


[152]

Walker HK, Hall WD, Hurst JW, Cabaniss CD. Creatine Kinase. Clinical Methods: The History, Physical, and Laboratory Examinations. 1990:():     [PubMed PMID: 21250193]


[153]

Kim EY, Lee JW, Suh MR, Choi WA, Kang SW, Oh HJ. Correlation of Serum Creatine Kinase Level With Pulmonary Function in Duchenne Muscular Dystrophy. Annals of rehabilitation medicine. 2017 Apr:41(2):306-312. doi: 10.5535/arm.2017.41.2.306. Epub 2017 Apr 27     [PubMed PMID: 28503465]


[154]

Kalyani RR, Corriere M, Ferrucci L. Age-related and disease-related muscle loss: the effect of diabetes, obesity, and other diseases. The lancet. Diabetes & endocrinology. 2014 Oct:2(10):819-29. doi: 10.1016/S2213-8587(14)70034-8. Epub 2014 Mar 6     [PubMed PMID: 24731660]


[155]

Drummond LM. Creatine phosphokinase levels in the newborn and their use in screening for Duchenne muscular dystrophy. Archives of disease in childhood. 1979 May:54(5):362-6     [PubMed PMID: 475411]


[156]

Percy ME, Chang LS, Murphy EG, Oss I, Verellen-Dumoulin C, Thompson MW. Serum creatine kinase and pyruvate kinase in Duchenne muscular dystrophy carrier detection. Muscle & nerve. 1979 Sep-Oct:2(5):329-39     [PubMed PMID: 492209]


[157]

Adam MP, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, Bonne G, Leturcq F, Ben Yaou R. Emery-Dreifuss Muscular Dystrophy. GeneReviews(®). 1993:():     [PubMed PMID: 20301609]


[158]

Takeshima K, Ariyasu H, Ishibashi T, Kawai S, Uraki S, Koh J, Ito H, Akamizu T. Myotonic dystrophy type 1 with diabetes mellitus, mixed hypogonadism and adrenal insufficiency. Endocrinology, diabetes & metabolism case reports. 2018:2018():. doi: 10.1530/EDM-17-0143. Epub 2018 Jan 18     [PubMed PMID: 29367875]

Level 3 (low-level) evidence

[159]

Kim HK, Lindquist DM, Serai SD, Mariappan YK, Wang LL, Merrow AC, McGee KP, Ehman RL, Laor T. Magnetic resonance imaging of pediatric muscular disorders: recent advances and clinical applications. Radiologic clinics of North America. 2013 Jul:51(4):721-42. doi: 10.1016/j.rcl.2013.03.002. Epub     [PubMed PMID: 23830795]

Level 3 (low-level) evidence

[160]

Wong CK, Gidali A, Harris V. Deformity or dysfunction? Osteopathic manipulation of the idiopathic cavus foot: A clinical suggestion. North American journal of sports physical therapy : NAJSPT. 2010 Feb:5(1):27-32     [PubMed PMID: 21509155]


[161]

Ilaslan H, Wenger DE, Shives TC, Unni KK. Unilateral hypertrophy of tensor fascia lata: a soft tissue tumor simulator. Skeletal radiology. 2003 Nov:32(11):628-32     [PubMed PMID: 14586575]


[162]

Grimm T, Kress W, Meng G, Müller CR. Risk assessment and genetic counseling in families with Duchenne muscular dystrophy. Acta myologica : myopathies and cardiomyopathies : official journal of the Mediterranean Society of Myology. 2012 Dec:31(3):179-83     [PubMed PMID: 23620649]


[163]

Nakamura A. Mutation-Based Therapeutic Strategies for Duchenne Muscular Dystrophy: From Genetic Diagnosis to Therapy. Journal of personalized medicine. 2019 Mar 4:9(1):. doi: 10.3390/jpm9010016. Epub 2019 Mar 4     [PubMed PMID: 30836656]


[164]

Pastore CA, Samesima N, Pereira-Filho HG. III SBC Guidelines on the Analysis and Issuance of Electrocardiographic Reports - Executive Summary. Arquivos brasileiros de cardiologia. 2016 Nov:107(5):392-402. doi: 10.5935/abc.20160173. Epub     [PubMed PMID: 27982266]


[165]

Sarubbi B, Li W, Somerville J. QRS width in right bundle branch block. Accuracy and reproducibility of manual measurement. International journal of cardiology. 2000 Aug:75(1):71-4     [PubMed PMID: 11054509]


[166]

Nikolić G. Dominant R wave in lead V1. Heart & lung : the journal of critical care. 1998 Sep-Oct:27(5):352-3     [PubMed PMID: 9777382]


[167]

Thrush PT, Allen HD, Viollet L, Mendell JR. Re-examination of the electrocardiogram in boys with Duchenne muscular dystrophy and correlation with its dilated cardiomyopathy. The American journal of cardiology. 2009 Jan 15:103(2):262-5. doi: 10.1016/j.amjcard.2008.08.064. Epub 2008 Oct 30     [PubMed PMID: 19121448]


[168]

Koene RJ, Adkisson WO, Benditt DG. Syncope and the risk of sudden cardiac death: Evaluation, management, and prevention. Journal of arrhythmia. 2017 Dec:33(6):533-544. doi: 10.1016/j.joa.2017.07.005. Epub 2017 Sep 1     [PubMed PMID: 29255498]


[169]

Groh WJ, Groh MR, Saha C, Kincaid JC, Simmons Z, Ciafaloni E, Pourmand R, Otten RF, Bhakta D, Nair GV, Marashdeh MM, Zipes DP, Pascuzzi RM. Electrocardiographic abnormalities and sudden death in myotonic dystrophy type 1. The New England journal of medicine. 2008 Jun 19:358(25):2688-97. doi: 10.1056/NEJMoa062800. Epub     [PubMed PMID: 18565861]


[170]

Segawa K, Komaki H, Mori-Yoshimura M, Oya Y, Kimura K, Tachimori H, Kato N, Sasaki M, Takahashi Y. Cardiac conduction disturbances and aging in patients with Duchenne muscular dystrophy. Medicine. 2017 Oct:96(42):e8335. doi: 10.1097/MD.0000000000008335. Epub     [PubMed PMID: 29049249]


[171]

Paganoni S, Amato A. Electrodiagnostic evaluation of myopathies. Physical medicine and rehabilitation clinics of North America. 2013 Feb:24(1):193-207. doi: 10.1016/j.pmr.2012.08.017. Epub 2012 Oct 16     [PubMed PMID: 23177039]


[172]

Cannon SC. Channelopathies of skeletal muscle excitability. Comprehensive Physiology. 2015 Apr:5(2):761-90. doi: 10.1002/cphy.c140062. Epub     [PubMed PMID: 25880512]


[173]

Roberts RG. Direct diagnosis of carriers of Duchenne and Becker muscular dystrophy by amplification of lymphocyte RNA. Lancet (London, England). 1991 Feb 23:337(8739):504     [PubMed PMID: 1704092]


[174]

El Sherif RM, Fahmy NA, Nonaka I, Etribi MA. Patterns of dystrophin gene deletion in Egyptian Duchenne/Becker muscular dystrophy patients. Acta myologica : myopathies and cardiomyopathies : official journal of the Mediterranean Society of Myology. 2007 Dec:26(3):145-50     [PubMed PMID: 18646563]


[175]

Sbiti A, El Kerch F, Sefiani A. Analysis of Dystrophin Gene Deletions by Multiplex PCR in Moroccan Patients. Journal of biomedicine & biotechnology. 2002:2(3):158-160     [PubMed PMID: 12488581]


[176]

Abbs S, Roberts RG, Mathew CG, Bentley DR, Bobrow M. Accurate assessment of intragenic recombination frequency within the Duchenne muscular dystrophy gene. Genomics. 1990 Aug:7(4):602-6     [PubMed PMID: 1974880]


[177]

Barresi R. From proteins to genes: immunoanalysis in the diagnosis of muscular dystrophies. Skeletal muscle. 2011 Jun 24:1(1):24. doi: 10.1186/2044-5040-1-24. Epub 2011 Jun 24     [PubMed PMID: 21798100]


[178]

Oliveira AS, Gabbai AA, Schmidt B, Kiyomoto BH, Lima JG, Minetti C, Bonilla E. Carrier detection of Duchenne and Becker muscular dystrophy using muscle dystrophin immunohistochemistry. Arquivos de neuro-psiquiatria. 1992 Dec:50(4):478-85     [PubMed PMID: 1309152]


[179]

Le Thanh P, Meinke P, Korfali N, Srsen V, Robson MI, Wehnert M, Schoser B, Sewry CA, Schirmer EC. Immunohistochemistry on a panel of Emery-Dreifuss muscular dystrophy samples reveals nuclear envelope proteins as inconsistent markers for pathology. Neuromuscular disorders : NMD. 2017 Apr:27(4):338-351. doi: 10.1016/j.nmd.2016.12.003. Epub 2016 Dec 21     [PubMed PMID: 28214269]


[180]

Bertrand AT, Bönnemann CG, Bonne G, FHL1 myopathy consortium. 199th ENMC international workshop: FHL1 related myopathies, June 7-9, 2013, Naarden, The Netherlands. Neuromuscular disorders : NMD. 2014 May:24(5):453-62. doi: 10.1016/j.nmd.2014.02.002. Epub 2014 Feb 14     [PubMed PMID: 24613424]


[181]

Folker ES, Baylies MK. Nuclear positioning in muscle development and disease. Frontiers in physiology. 2013 Dec 12:4():363. doi: 10.3389/fphys.2013.00363. Epub 2013 Dec 12     [PubMed PMID: 24376424]


[182]

Peverelli L, Testolin S, Villa L, D'Amico A, Petrini S, Favero C, Magri F, Morandi L, Mora M, Mongini T, Bertini E, Sciacco M, Comi GP, Moggio M. Histologic muscular history in steroid-treated and untreated patients with Duchenne dystrophy. Neurology. 2015 Nov 24:85(21):1886-93. doi: 10.1212/WNL.0000000000002147. Epub 2015 Oct 23     [PubMed PMID: 26497992]


[183]

Angelini C, Tasca E. Fatigue in muscular dystrophies. Neuromuscular disorders : NMD. 2012 Dec:22 Suppl 3(3-3):S214-20. doi: 10.1016/j.nmd.2012.10.010. Epub     [PubMed PMID: 23182642]


[184]

Guilleminault C, Philip P, Robinson A. Sleep and neuromuscular disease: bilevel positive airway pressure by nasal mask as a treatment for sleep disordered breathing in patients with neuromuscular disease. Journal of neurology, neurosurgery, and psychiatry. 1998 Aug:65(2):225-32     [PubMed PMID: 9703177]


[185]

Reardon W, MacMillan JC, Myring J, Harley HG, Rundle SA, Beck L, Harper PS, Shaw DJ. Cataract and myotonic dystrophy: the role of molecular diagnosis. The British journal of ophthalmology. 1993 Sep:77(9):579-83     [PubMed PMID: 8218057]


[186]

Adam MP, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, Darras BT, Urion DK, Ghosh PS. Dystrophinopathies. GeneReviews(®). 1993:():     [PubMed PMID: 20301298]


[187]

Palladino A, D'Ambrosio P, Papa AA, Petillo R, Orsini C, Scutifero M, Nigro G, Politano L. Management of cardiac involvement in muscular dystrophies: paediatric versus adult forms. Acta myologica : myopathies and cardiomyopathies : official journal of the Mediterranean Society of Myology. 2016 Dec:35(3):128-134     [PubMed PMID: 28484313]


[188]

Tsao CY, Mendell JR. Coexisting muscular dystrophies and epilepsy in children. Journal of child neurology. 2006 Feb:21(2):148-50     [PubMed PMID: 16566880]


[189]

Foff EP, Mahadevan MS. Therapeutics development in myotonic dystrophy type 1. Muscle & nerve. 2011 Aug:44(2):160-9. doi: 10.1002/mus.22090. Epub 2011 May 23     [PubMed PMID: 21607985]


[190]

Groh WJ. Mexiletine is an effective antimyotonia treatment in myotonic dystrophy type 1. Neurology. 2011 Jan 25:76(4):409; author reply 409. doi: 10.1212/WNL.0b013e3181fe72d7. Epub     [PubMed PMID: 21263144]


[191]

Stunnenberg BC, Woertman W, Raaphorst J, Statland JM, Griggs RC, Timmermans J, Saris CG, Schouwenberg BJ, Groenewoud HM, Stegeman DF, van Engelen BG, Drost G, van der Wilt GJ. Combined N-of-1 trials to investigate mexiletine in non-dystrophic myotonia using a Bayesian approach; study rationale and protocol. BMC neurology. 2015 Mar 25:15():43. doi: 10.1186/s12883-015-0294-4. Epub 2015 Mar 25     [PubMed PMID: 25880166]


[192]

Eccleston C, Cooper TE, Fisher E, Anderson B, Wilkinson NM. Non-steroidal anti-inflammatory drugs (NSAIDs) for chronic non-cancer pain in children and adolescents. The Cochrane database of systematic reviews. 2017 Aug 2:8(8):CD012537. doi: 10.1002/14651858.CD012537.pub2. Epub 2017 Aug 2     [PubMed PMID: 28770976]

Level 1 (high-level) evidence

[193]

Topaloglu H, Gloss D, Moxley RT 3rd, Ashwal S, Oskoui M. Practice guideline update summary: Corticosteroid treatment of Duchenne muscular dystrophy: Report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology. 2016 Jul 12:87(2):238. doi: 10.1212/01.wnl.0000489553.99227.18. Epub     [PubMed PMID: 27402942]

Level 1 (high-level) evidence

[194]

Angelini C, Peterle E. Old and new therapeutic developments in steroid treatment in Duchenne muscular dystrophy. Acta myologica : myopathies and cardiomyopathies : official journal of the Mediterranean Society of Myology. 2012 May:31(1):9-15     [PubMed PMID: 22655511]


[195]

Guglieri M, Bushby K, McDermott MP, Hart KA, Tawil R, Martens WB, Herr BE, McColl E, Wilkinson J, Kirschner J, King WM, Eagle M, Brown MW, Willis T, Hirtz D, Shieh PB, Straub V, Childs AM, Ciafaloni E, Butterfield RJ, Horrocks I, Spinty S, Flanigan KM, Kuntz NL, Baranello G, Roper H, Morrison L, Mah JK, Manzur AY, McDonald CM, Schara U, von der Hagen M, Barohn RJ, Campbell C, Darras BT, Finkel RS, Vita G, Hughes I, Mongini T, Pegoraro E, Wicklund M, Wilichowski E, Bryan Burnette W, Howard JF, McMillan HJ, Thangarajh M, Griggs RC. Developing standardized corticosteroid treatment for Duchenne muscular dystrophy. Contemporary clinical trials. 2017 Jul:58():34-39. doi: 10.1016/j.cct.2017.04.008. Epub 2017 Apr 24     [PubMed PMID: 28450193]


[196]

Leung DG, Wagner KR. Therapeutic advances in muscular dystrophy. Annals of neurology. 2013 Sep:74(3):404-11. doi: 10.1002/ana.23989. Epub     [PubMed PMID: 23939629]

Level 3 (low-level) evidence

[197]

Adam MP, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, Preston MK, Tawil R, Wang LH. Facioscapulohumeral Muscular Dystrophy. GeneReviews(®). 1993:():     [PubMed PMID: 20301616]


[198]

Day JW, Ricker K, Jacobsen JF, Rasmussen LJ, Dick KA, Kress W, Schneider C, Koch MC, Beilman GJ, Harrison AR, Dalton JC, Ranum LP. Myotonic dystrophy type 2: molecular, diagnostic and clinical spectrum. Neurology. 2003 Feb 25:60(4):657-64     [PubMed PMID: 12601109]


[199]

Lim KRQ, Yokota T. Invention and Early History of Exon Skipping and Splice Modulation. Methods in molecular biology (Clifton, N.J.). 2018:1828():3-30. doi: 10.1007/978-1-4939-8651-4_1. Epub     [PubMed PMID: 30171532]


[200]

Nguyen Q, Yokota T. Immortalized Muscle Cell Model to Test the Exon Skipping Efficacy for Duchenne Muscular Dystrophy. Journal of personalized medicine. 2017 Oct 16:7(4):. doi: 10.3390/jpm7040013. Epub 2017 Oct 16     [PubMed PMID: 29035327]


[201]

Sussman M. Duchenne muscular dystrophy. The Journal of the American Academy of Orthopaedic Surgeons. 2002 Mar-Apr:10(2):138-51     [PubMed PMID: 11929208]


[202]

Ciudin RN, Dragatoiu NC, Sipos S, Tesloianu DN, Ursaru AM, Brezeanu R, Coman IM. Fast Progressing His-Purkinje Conduction Disturbances in a Myotonic Dystrophy Pacient. Maedica. 2018 Jun:13(2):152-154. doi: 10.26574/maedica.2018.13.2.152. Epub     [PubMed PMID: 30069244]


[203]

Altekin RE, Yanikoglu A, Ucar M, Ermis C. Complete AV block and cardiac syncope in a patient with Duchenne muscular dystrophy. Journal of cardiology cases. 2011 Apr:3(2):e68-e70. doi: 10.1016/j.jccase.2011.01.004. Epub 2011 Feb 16     [PubMed PMID: 30532840]

Level 3 (low-level) evidence

[204]

Copeland SA, Levy O, Warner GC, Dodenhoff RM. The shoulder in patients with muscular dystrophy. Clinical orthopaedics and related research. 1999 Nov:(368):80-91     [PubMed PMID: 10613155]


[205]

Shin J, Tajrishi MM, Ogura Y, Kumar A. Wasting mechanisms in muscular dystrophy. The international journal of biochemistry & cell biology. 2013 Oct:45(10):2266-79. doi: 10.1016/j.biocel.2013.05.001. Epub 2013 May 11     [PubMed PMID: 23669245]


[206]

de Souza MA, Figueiredo MM, de Baptista CR, Aldaves RD, Mattiello-Sverzut AC. Beneficial effects of ankle-foot orthosis daytime use on the gait of Duchenne muscular dystrophy patients. Clinical biomechanics (Bristol, Avon). 2016 Jun:35():102-10. doi: 10.1016/j.clinbiomech.2016.04.005. Epub 2016 Apr 22     [PubMed PMID: 27139255]


[207]

Gupta A, Nalini A, Arya SP, Vengalil S, Khanna M, Krishnan R, Taly AB. Ankle-Foot Orthosis in Duchenne Muscular Dystrophy: A 4 year Experience in a Multidisciplinary Neuromuscular Disorders Clinic. Indian journal of pediatrics. 2017 Mar:84(3):211-215. doi: 10.1007/s12098-016-2251-7. Epub 2016 Nov 5     [PubMed PMID: 27815810]


[208]

Olivé M, Kley RA, Goldfarb LG. Myofibrillar myopathies: new developments. Current opinion in neurology. 2013 Oct:26(5):527-35. doi: 10.1097/WCO.0b013e328364d6b1. Epub     [PubMed PMID: 23995273]

Level 3 (low-level) evidence

[209]

Nigro V. Improving the course of muscular dystrophy? Acta myologica : myopathies and cardiomyopathies : official journal of the Mediterranean Society of Myology. 2012 Oct:31(2):109     [PubMed PMID: 23097600]


[210]

Read AP, Donnai D. What can be offered to couples at (possibly) increased genetic risk? Journal of community genetics. 2012 Jul:3(3):167-74. doi: 10.1007/s12687-012-0105-1. Epub 2012 Jul 4     [PubMed PMID: 22760671]


[211]

Chawla J. Stepwise approach to myopathy in systemic disease. Frontiers in neurology. 2011:2():49. doi: 10.3389/fneur.2011.00049. Epub 2011 Aug 5     [PubMed PMID: 21886637]


[212]

Passamano L, Taglia A, Palladino A, Viggiano E, D'Ambrosio P, Scutifero M, Rosaria Cecio M, Torre V, DE Luca F, Picillo E, Paciello O, Piluso G, Nigro G, Politano L. Improvement of survival in Duchenne Muscular Dystrophy: retrospective analysis of 835 patients. Acta myologica : myopathies and cardiomyopathies : official journal of the Mediterranean Society of Myology. 2012 Oct:31(2):121-5     [PubMed PMID: 23097603]

Level 2 (mid-level) evidence

[213]

Bertini E, D'Amico A, Gualandi F, Petrini S. Congenital muscular dystrophies: a brief review. Seminars in pediatric neurology. 2011 Dec:18(4):277-88. doi: 10.1016/j.spen.2011.10.010. Epub     [PubMed PMID: 22172424]


[214]

Birnkrant DJ, Bushby K, Bann CM, Alman BA, Apkon SD, Blackwell A, Case LE, Cripe L, Hadjiyannakis S, Olson AK, Sheehan DW, Bolen J, Weber DR, Ward LM, DMD Care Considerations Working Group. Diagnosis and management of Duchenne muscular dystrophy, part 2: respiratory, cardiac, bone health, and orthopaedic management. The Lancet. Neurology. 2018 Apr:17(4):347-361. doi: 10.1016/S1474-4422(18)30025-5. Epub 2018 Feb 3     [PubMed PMID: 29395990]

Level 3 (low-level) evidence