Sie sind auf Seite 1von 73

Chapter 216: The Muscular Dystrophies

PART 25: MUSCLE


Chapter 216: The Muscular Dystrophies
Peter B. Kang, Louis M. Kunkel
Abstract
1. The muscular dystrophies are a broad class of primary muscle disorders with a diverse range of
genetic etiologies, clinical presentations, and natural history. The disease generally arises through a
loss of function of the affected protein product. They share a common histopathologic picture of
muscle degeneration and regeneration, characterized by fiber necrosis, inflammation, and
fibroadipose replacement of muscle fibers. This distinguishes them from the congenital myopathies,
which are associated with muscle fiber dysmorphology, often due to abnormal accumulations of
various proteins.
2. The first description of Duchenne muscular dystrophy (DMD) (OMIM 310200), the most common
form, is generally attributed to Duchenne de Boulogne in 1861, although there are several reports
published earlier in the nineteenth century by Edward Meryon and others that appear to present the
same disease. A major milestone in the genetics of this disorder came in 1986, when the gene
causing DMD was cloned. A year later, its protein product, dystrophin, was identified. Since then,
several dozen genes have been associated with various forms of muscular dystrophy (gene
sequences available at http://www.dmd.nl/).
3. The complexities of the genotype-phenotype correlations in the muscular dystrophies make it difficult
to classify them. Two different schemas are presented, one organized by categories of the various
proteins encoded by the causative genes, the other organized by phenotype. The former will be more
useful for scientific researchers, the latter for clinicians.
4. In a phenotypic classification, the X-linked dystrophinopathies DMD and Becker muscular dystrophy
(BMD) (OMIM 300376) are the most common forms of muscular dystrophy. Both are caused by
mutations in the gene DMD (also known as dystrophin). The classic form of BMD has a milder
phenotype than DMD, but the boundary between the two phenotypes is not always easy to
determine, as there is a spectrum of clinical manifestations.
5. The autosomal dominant limb girdle muscular dystrophies (LGMDs) are to some extent
phenotypically similar to the milder forms of dystrophinopathy. There are three autosomal dominant
LGMDs for which causative genes have been identified. LGMD1A (myotilinopathy) (OMIM 159000) is
caused by mutations in TTID. Mutations in LMNA cause a variety of phenotypes, including LGMD1B
(laminopathy) (OMIM 159001), Emery-Dreifuss muscular dystrophy (OMIM 310300, 181350,
604929), cardiomyopathy, lipodystrophy, and Hutchinson-Gilford progeria. LGMD1C is also known as
caveolinopathy (OMIM 607801) and is caused by mutations in CAV3. There are three other forms of
autosomal dominant LGMD for which causative genes have not yet been identified: LGMD1D,
LGMD1E, and LGMD1F.
6. The autosomal recessive LGMDs tend to be more severe, and have some phenotypic similarities to
the more severe end of dystrophinopathy spectrum. They include LGMD2A (calpainopathy; CAPN)
(OMIM 253600), LGMD2B (dysferlinopathy; DYSF) (OMIM 253601), LGMD2C (-sarcoglycanopathy;
SGCG) (OMIM 253700), LGMD2D (-sarcoglycanopathy; SGCA) (OMIM 608099), LGMD2E
(-sarcoglycanopathy; SGCB) (OMIM 604286), LGMD2F (-sarcoglycanopathy; SGCD) (OMIM
601287), LGMD2G (telethoninopathy; TCAP) (OMIM 601954), LGMD2H (TRIM32) (OMIM 254110),

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

-1-

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

7.

8.

9.

10.

11.

12.

13.

14.

LGMD2I (FKRP) (OMIM 607155), LGMD2J (TTN) (OMIM 608807), and LGMD2K (POMT1) (OMIM
609308).
Congenital muscular dystrophies form a third category of muscular dystrophies after the
dystrophinopathies and the LGMDs. They include milder phenotypes such as Ullrich disease
(COL6A1, COL6A2, COL6A3) (OMIM 254090) and rigid spine syndrome (SEPN1) (OMIM 602771).
The most common congenital muscular dystrophy, traditionally referred to as merosin deficiency (MDC1A;
LAMA2) (OMIM 607855), has a moderately severe phenotype. A mutation in FKRP may cause a
moderate form of congenital muscular dystrophy (MDC1C) (OMIM 606612) as well as LGMD2I. The three
classic severe congenital muscular dystrophies are Fukuyama congenital muscular dystrophy
(FCMD) (OMIM 253800), muscle-eye-brain disease (POMGnT1) (OMIM 253280), and
Walker-Warburg syndrome (POMT1, FCMD, FKRP) (OMIM 236670).
Some distal myopathies have dystrophic features on histology, including Miyoshi myopathy (DYSF)
(OMIM 254130), which is allelic to LGMD2B; and tibial muscular dystrophy (Udd myopathy; TTN)
(OMIM 600334), which is allelic to LGMD2J.
Several unique phenotypes do not easily fit into one of the above categories, including Bethlem
myopathy (COL6A1, COL6A2, COL6A3) (OMIM 158810), Emery-Dreifuss muscular dystrophy (EMD,
LMNA) (OMIM 310300, 181350, 604929), facioscapulohumeral muscular dystrophy (OMIM 158900),
and oculopharyngeal muscular dystrophy (PABP2) (OMIM 164300).
The natural history ranges widely in these disorders, from the most severe congenital muscular
dystrophies that are rapidly fatal to very mild diseases that have onset in adulthood and do not cause
significant disabilities. Other organ systems may be involved, depending on the specific disease, including
the heart, lungs, spine, eyes, and brain. In the more severe forms, the usual cause of death is a cardiac or
pulmonary complication.
The protein products of the genes known to cause muscular dystrophy perform a variety of functions
and are clustered in several specific locations within or outside the muscle fiber. One cluster forms the
dystrophin-associated protein complex (DAPC), which is linked to a second cluster in the extracellular
matrix complex through sarcolemmal proteins. Three other clusters lie in the sarcomere, Golgi apparatus,
and nucleus. There are a few proteins that do not easily fall into these five main categories, such as
enzymes. The functions range from structural to signaling to glycosylation, depending on the specific
protein. Many of these proteins are also expressed in nonmuscle tissues, often leading to multiorgan
involvement in the muscular dystrophies.
The diagnosis of the muscular dystrophies has changed dramatically since the gene for DMD was
cloned in 1986. Many children with DMD and BMD no longer require uncomfortable and invasive
diagnostic tests such as electromyography and muscle biopsy. In about two-thirds of cases, deletion and
duplication analyses on DNA obtained from blood lymphocytes suffice to confirm the diagnosis after an
elevated creatine kinase (CK) level has been documented. Muscle biopsy may still be required in
many of the remaining cases, as well as in many of the other forms of muscular dystrophy, although
mutation analysis is increasingly being used to confirm the diagnosis.
Many of the genes that cause the muscular dystrophies have homologues in other organisms. Three
organisms that are commonly used as models of muscular dystrophy are the mouse, dog, and zebrafish.
The mdx mouse is the most widely used model. The xmd dog more closely replicates humans in
general and DMD in particular compared with the mdx mouse, but is a more difficult organism to work
with. Zebrafish models of muscular dystrophy have recently been developed and show promise in certain
settings due to their rapid reproductive cycle.
Significant progress has been made in the therapy of the muscular dystrophies over the past two
decades. Steroid therapy prolongs ambulation in DMD. Surgery for scoliosis alleviates the respiratory
complications and makes the patient more comfortable. The development of portable respirators has
preserved motility late in the course even when respiratory failure occurs. Angiotensin-converting
enzyme (ACE) inhibitors improve the course of cardiomyopathy, which complicates DMD, BMD, and
other muscular dystrophies. Physical and occupational therapy also have been helpful. The life
expectancy and quality of life have both improved significantly for affected individuals. A definitive cure,

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

-2-

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

however, remains elusive. A new generation of potential therapies has emerged from the laboratory, and
hope remains that one of these will eventually develop into a cure.

Background
History
Several works of art dating as far back as ancient Egypt, as well as some medieval paintings, are
suspected of portraying undiagnosed cases of muscular dystrophy (Emery, 1995). In the nineteenth
century, Sir Charles Bell, Gaetano Conte, Richard Partridge, William Little, and Edward Meryon all
published descriptions of what most likely were cases of muscular dystrophy (Emery, 1995; Little, 1953;
Meryon, 1852; Tyler, 2003). These descriptions were not, however, recognized at the times of publication
as representing a distinct disease.
Guillaume Benjamin Amand Duchenne de Boulogne, a French physician, published a description of his
eponymous disorder in 1861 (Duchenne, 1861). His first case was that of a 9-year-old boy who began
walking at 18 months. The patient had difficulty walking and getting up after falling, and had equinovarus
deformity. Duchenne noted pseudohypertrophy of the calves and mental retardation. He studied this
disorder in more detail and described a number of other cases, thus leading to its recognition as a disease
entity (Duchenne, 1868).
William Gowers defined the natural history in great detail and described what is now known as the Gowers
sign (Gowers, 1879). He noted the onset before age 6, loss of ambulation at 10 to 12 years, and death by
14 to 18 years (Emery, 1995). Additionally, he accurately described the patterns of muscle weakness,
wasting, and pseudohypertrophy.
Facioscapulohumeral muscular dystrophy was described later in the nineteenth century. For most of the
twentieth century, advances were restricted to descriptions of other forms of muscular dystrophy, including
BMD (Becker, 1953), limb girdle muscular dystrophy (LGMD), Emery-Dreifuss muscular dystrophy,
oculopharyngeal muscular dystrophies, and congenital muscular dystrophies.
The modern history of the muscular dystrophies began in the 1980s (Kunkel, 2005). The hunt for the gene
responsible for DMD had one important advantage and one important handicap. The advantage was that
the X-linked inheritance limited the search to the X chromosome. The handicap was that the biochemical
defect was also unknown. This great intellectual effort by many groups around the world was conducted at
a time when the polymerase chain reaction (PCR) was in its infancy and DNA sequencing was a laborious
process involving copious use of radioactive markers rather than the fluorescent ones involved in current
automated DNA sequencers.
The first step was the creation of a recombinant DNA library for the X chromosome. Chromosomes were
isolated from cell lines obtained from patients with 48, XXXX (Davies et al, 1981) and 49, XXXXY (Kunkel
et al, 1982) aneuploidy. The X chromosomes were sorted from the other chromosomes based on their
prominent fluorescence peaks on flow cytometry. After restriction endonuclease digestion, the resulting
DNA fragments were cloned into phage libraries.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

-3-

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

The gene was localized to the short arm of the X chromosome using two independent approaches:
linkage analysis of restriction fragment length polymorphisms in patients with DMD (Davies et al, 1983;
Murray et al, 1982) and BMD (Brown et al, 1985; Kingston et al, 1984); and mapping of X-autosome
reciprocal translocations in girls with DMD (Boyd et al, 1986; Verellen-Dumoulin et al, 1984). The former
led the way to accurate prenatal diagnosis of DMD, even though the responsible gene had yet to be
identified (Bakker et al, 1985). One of the X-autosome reciprocal translocation cases involved the transfer
of part of a ribosomal DNA repeat unit from chromosome 21 to the X chromosome, creating a site on the
X chromosome near the DMD gene that could be explored with rDNA probes (Worton et al, 1984).
Intense investigation of another unusual patient, B.B., also was underway by several groups. This boy had
four X-linked disorders: DMD, retinitis pigmentosa, chronic granulomatous disease, and the rare red-cell
phenotype McLeod. Karyotyping suggested a deletion in the Xp21 region of the X chromosome, while
Southern blot analysis of his DNA demonstrated a lack of hybridization of the 754 probe, confirming the
deletion and also excluding the possibility that the missing DNA had been translocated to an autosome
(Francke et al, 1985).
Further characterization of B.B.s deletion was accomplished using a hybridization method called
phenol-enhanced reassociation technique (PERT). A small amount of DNA isolated from a 48, XXXX
individual was cleaved with MboI, leaving ends compatible with cloning into BamH1 sites. These cleaved
control samples were then mixed with a much larger amount of B.B.s DNA, which had been sheared to
create blunt ends. Thus, only those restriction fragments from the control DNA that could not hybridize to
B.B.s DNA would self-hybridize and be able to insert themselves into a vector cleaved with BamH1.
There were 125 clones isolated in all, but only 4 were confirmed to be absent in B.B.s DNA by Southern
blot analysis; they were named pERT55, pERT145, pERT84, and pERT87 (Kunkel et al, 1985). Another
set of clones was isolated using the rDNA probes mentioned above (Ray et al, 1985).
One clone from each group failed to hybridize with the DNA of a subset of DMD and BMD patients,
suggesting that these clones corresponded to parts of the causative gene. These clones were named
DXS164 (pERT87) (Kunkel, 1986; Monaco et al, 1985) and DXS206 (XJ-1.1) (Ray et al, 1985).
Chromosome walking was used to isolate large blocks of DNA surrounding the original deletion-detecting
clones and exons identified by sequence conservation. Similar chromosome walks were undertaken at the
DXS206 (XJ-1.1) locus and again sequence conservation used to identify putative exons. One of these
putative exons hybridized to a large RNA species obtained from human fetal skeletal muscle. This
fragment was used to isolate cDNA clones representing at least eight small regions, presumably exons, in
the DXS164 locus (Monaco et al, 1986). These turned out to be the first exons sequenced for the gene
deficient in DMD. This was soon followed by another clone containing further independent exons of a
possibly very large gene (Burghes et al, 1987).
Subsequently, the complete sequence of the predicted protein product of the DMD gene at Xp21 was
obtained (Koenig et al, 1987) and the protein product identified and named dystrophin (Hoffman et al,
1987). Dystrophin was then localized to the subsarcolemmal region (Arahata et al, 1988; Bonilla et al,
1988; Watkins et al, 1988; Zubrzycka-Gaarn et al, 1988). In the two decades since these discoveries,
several dozen other genes associated with less common forms of muscular dystrophy have been cloned
(Fig. 216-1). Further details about these specific genes and their protein products are provided below. The
diagnostic evaluation of children with muscular dystrophy has been revolutionized as a result.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

-4-

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

-5-

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

The development of knowledge of muscle fiber structure over time, including the dystrophin-associated
protein complex. A, Muscle fiber structure before the 1980s. B, The late 1980s to early 1990s, when
dystrophin and the dystroglycans were characterized. C, By the mid-1990s, the sarcoglycan complex and
several further proteins were identified. D, By the late 1990s, dysferlin and caveolin were identified, as
well as fukutin in the Golgi compl...
Classification
The muscular dystrophies are most commonly classified phenotypically (Table 216-1). However, when
thinking about the muscular dystrophies on a molecular level, it is most useful to categorize them by the
cellular location of the encoded protein in relation to the muscle fiber (Table 216-2). The clinical
presentation and natural history section will be organized according to the categories listed in Table
216-1, while the genetics and biochemistry section will be organized according to the listings in Table
216-2.
Table 216-1: Phenotypic Classification of the Muscular Dystrophies

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

-6-

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Clinical Form

Abbreviation

inheritance

Key Features

Duchenne (OMIM No.


310200)

DMD

X-linked

Severe, progressive muscle


degeneration, onset ages 46, loss of
ambulation at ages 912, death at ages
1420 of respiratory
failure/cardiomyopathy

Becker (OMIM No.


310200)

BMD

X-linked

Mild form, onset after age 16, muscle


pain, dilated cardiomyopathy may
require heart transplant

Limb-Girdle (OMIM: see


Table 216-2)

LGMD

AD or AR

Severity variable, overlap DMD and


BMD, involves primarily shoulder and
pelvic girdle muscles

Congenital (classic)
(OMIM 156225)

CMD

AR

Hypotonia, joint contractures, general


muscle weakness, many never become
ambulatory

Congential (Fukuyama)
(OMIM 253800)

FMD

AR

Common only in Japan, similar to


classic form but severe mental
impairment, CNS alterations

Emery-Dreifuss, (OMIM
310300)

EDMD

X-linked

Contractures of elbow, neck, and spine,


scapulohumeroperoneal weakness,
cardiac conduction defect

Oculopharyngeal (OMIM:
see Table 216-2)

OPMG

AD or AR

Eyelid ptosis and dysphagia, onset after


age 50,

Fascioscapulohumeral
(OMIM, 158900)

FSHMD

AD

Facial and shoulder weakness, scapular


winging, variable cardiac involvement

NOTE: AR = autosomal recessive; AD = autosomal dominant.

Table 216-2: Cellular Localization of Proteins Involved in the Muscular Dystrophies

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

-7-

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Disease

Chromosome
Location

Defective
Protein

OMIM

Genbank
Accession No.

DMD

Xp21

Dystrophin

310200

NM000109

BMD

Xp21

Dystrophin

310200

NM000109

XLDC

Xp21

Dystrophin

310200

NM000109

LGMD-1A

5q22-34

Unknown

159000

LGMD-1B

1q11-21

Unknown

159001

LGMD-1C

3p25

Caveolin-3

601253

NM001234

LGMD-2A (Amish)

15q15-21

Calpain-3

253600

AF127765

114240
LGMD-2B/Mioshi
myopathy

2p13

Dysferlin

253601

NM003494

603009
LGMD-2C/SCARMD

13q12

-Sarcoglycan

253700

NM000231

LGMD-2D/ARMD

17q12-21

-Sarcoglycan

600119

NM000023

LGMD-2E
(Amish)/ARMD

4q12

-Sarcoglycan

600900

NM000232

LGMD-2F

5q33-34

-Sarcoglycan

601287

NM000337

601411
LGMD-2G

17q11-12

Unknown

601954

LGMD-2H (Hutterite)

9q31-34

Unknown

254110

CMD, classic

6q2

Merosin

156225

AI636560

CMD, Fukuyama

9q31-33

Fukutin

253800

AB008226

EDMD

Xq28

Emerin

310300

NM000117

OPMD

4q11-13

PABP2

164300

NM004643

257950
FSHMD

4q

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

Tandem repeat

-8-

158900

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Clinical Presentation and Natural History: Dystrophinopathies


Duchenne Muscular Dystrophy
The original descriptions of DMD as published by Meryon, Duchenne, Gowers, and their peers in the
nineteenth century remain unmatched. Nearly all affected individuals are male due to the X-linked
recessive inheritance; rare female patients are seen in cases of X-autosomal translocations. The onset of
symptoms is typically between the ages of 3 and 5 years, although some variation exists. In retrospect,
some parents recall slightly delayed early motor milestones, typically not significant enough to warrant
evaluation. The initial presenting symptoms may include frequent tripping and falling, toe-walking,
waddling gait, or vague complaints of difficulty walking. Occasionally an affected child is identified as a
result of complications of anesthesia, including cardiac arrest and rhabdomyolysis (Farrell, 1994; Larsen
et al, 1989).
The overall incidence has been estimated to be 1 in 3300 live male births. DMD is found throughout the
world and shows no geographic or ethnic predisposition.
The physical examination is notable for proximal motor weakness, most prominently at the hip girdle
muscles. Confrontation muscle strength testing is not always possible at this age, especially if there are
cognitive delays as well. Thus, the most reliable means of assessing proximal muscle strength remains
the Gowers sign, described in the late nineteenth century. The child is asked to rise from a sitting or lying
position to a standing position. A child with hip extensor weakness as is found in DMD will not be able to
rise directly to a standing position. In a classic Gowers sign, an affected child will compensate for weak
hip extensor muscles by raising the hips in the air first with legs extended, then "crawl" up the legs with
the hands to elevate the trunk. There are variations on this maneuver that may be used depending on the
extent of weakness and the personal preference of the child, but the constant finding is the elevation of
the hips higher than the head at some point during the maneuver. These other maneuvers are often
referred to as "modified Gowers signs."
An older child or a particularly cooperative younger one will be able to comply with confrontation testing,
demonstrating a predominantly proximal symmetrical pattern of weakness early in the course, with spread
to more distal muscles in later years. The legs are typically weaker than the arms. There are other,
somewhat puzzling patterns of weakness. The tibialis anterior, which controls dorsiflexion (upward
movement) of the foot, is affected earlier and more severely than the gastrocnemius and soleus, which
control plantarflexion (downward movement) of the foot. However, both are large calf muscles. There is no
immediate explanation for these patterns, which are consistently found in children with DMD.
The other major physical sign evident early in the course is muscle pseudohypertrophy, which is typically
most noticeable in the calves. However, it can be present in other muscles as well. The enlargement of
the muscles is referred to as pseudohypertrophy rather than hypertrophy because it is due to fibroadipose
accumulation rather than muscle fiber enlargement, and is a sign of weakness rather than strength. Deep
tendon reflexes are initially normal. The diagnostic evaluation of these children is described in detail
below.
The weakness itself is steadily progressive as demonstrated by sequential neurologic examinations
(Boland et al, 1996; Brooke et al, 1989), but the functional abilities most visible to families may decline in a
more stepwise fashion (Brooke et al, 1983). There may be an apparent improvement in motor skills
between 3 and 6 years, while the functional decline typically accelerates after 8 years (Brooke et al,
1983). Without prednisone therapy, an affected child loses ambulation at a mean age of about 10 years,

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

-9-

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

with a range typically from 7 to 13 years. With prednisone therapy, the mean age of loss of ambulation
may be delayed to 12 years (DeSilva et al, 1987). Further details on prednisone therapy may be found in
the therapy section.
Some affected individuals have essentially normal cognitive function, but a large number have mild static
impairments, measured to be on average one standard deviation below the mean, with especially
pronounced verbal impairments in many cases and emotional difficulties (Leibowitz, Dubowitz, 1981).
A major complication is scoliosis, which typically develops after the child begins to use a wheelchair
full-time. The scoliosis causes significant discomfort, social embarrassment, and functional difficulties. It
also can worsen respiratory function by decreasing the lung volume on one side, leading to diminished
vital capacity and atelectasis. The latter may increase the incidence of pneumonia.
Respiratory function is also compromised by diaphragm and intercostal muscle weakness. Pulmonary
complications form a leading cause of death, due to the combination of diminished vital capacity, weak
respiratory effort, and increased risk of pneumonias.
The other leading cause of death derives from cardiac complications, including cardiomyopathy and
arrhythmias (Brooke et al, 1989). The arrhythmias in particular are an unpredictable cause of death; fatal
rhythm disturbances typically occur later in the course but sometimes develop suddenly at an earlier age.
Without any treatment, the life expectancy rarely exceeds the adolescent years, and this was the
prognosis until the past several decades. A combination of improved diagnosis and therapies, as outlined
below, has lengthened the life expectancy significantly, such that many affected individuals now survive
past 30 years.
Becker Muscular Dystrophy
BMD is caused by the same gene as DMD; thus it is also X-linked recessive and almost always affects
males. The incidence has been estimated to be about 1 in 20,000 live male births. In contrast to DMD,
BMD patients typically have in-frame mutations, and thus some functional dystrophin protein is preserved
(Monaco et al, 1988). The phenotype is therefore milder, with onset usually in the second decade of life.
The range of severity is broad. Some patients lose the ability to walk independently during late
adolescence, while others remain ambulatory through most of adulthood. The cardiac complications are
the most serious ones; in fact, the cardiac manifestations may be the initial presentation of the condition in
some patients. Intelligence is usually normal.
Duchenne Muscular Dystrophy/Becker Muscular Dystrophy Intermediate Type
In some cases, boys with mutations in DMD will have a phenotype that is intermediate in age of onset and
severity between DMD and BMD. The prognosis also falls in the range between the two classic
diagnoses.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 10 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Duchenne Muscular Dystrophy/Becker Muscular Dystrophy Carriers


Female carriers of DMD and BMD mutations express, on average, 50% of the normal level of dystrophin.
This is enough to prevent clinical weakness in most of these individuals. However, about 8% of carriers
have mild skeletal muscle involvement (Politano et al, 1996), most likely due to skewed X-inactivation
(Matthews, Karpati, 1996). The role of X-inactivation has been demonstrated in monozygotic twin female
carriers with divergent phenotypes (Burn et al, 1986; Lupski et al, 1991; Richards et al, 1990). The more
serious issue is potential cardiomyopathy, which often occurs in the absence of skeletal muscle
manifestations. The incidence of clinically significant cardiac abnormalities has been estimated to be from
0 to 15% in female carriers under the age of 16 (Nolan et al, 2003; Politano et al, 1996), and 44.5% in
adult carriers females (Politano et al, 1996). Adult female carriers should definitely have cardiac
evaluations including electrocardiograms and echocardiograms as soon as they are diagnosed. It is more
difficult to determine whether female carriers should have cardiac evaluations in childhood. Certainly, this
should become a consideration if the child experiences potential cardiac symptoms (easy fatigability,
chest pain, dyspnea on exertion, palpitations) or plans to participate in a competitive athletic activity.
X-Linked Dilated Cardiomyopathy
Some cardiomyopathies are inherited in an X-linked manner with little or no skeletal muscle involvement
associated with mutations in DMD. Some of these mutations are located in the promoter and 5 end of the
gene (Milasin et al, 1996; Muntoni et al, 1993), while other mutations interfere with cardiac-specific splice
sites (Ferlini et al, 1998). Notably, cardiomyopathy caused by Coxsackie virus B3 is associated with
cleavage of cardiac dystrophin by enteroviral protease 2A (Badorff et al, 1999).
Clinical Presentation and Natural History: Limb Girdle Muscular Dystrophies (LGMD)
LGMDs are a large class of muscular dystrophies that are caused by genes other than DMD (dystrophin).
The phenotype ranges widely, from those with early onset and severe disability resembling that of DMD,
to milder phenotypes that more closely resemble BMD. Were it not for the difference in inheritance
patterns and historic distinctions in classification, DMD, BMD, and LGMD could all be considered part of a
large family of childhood- and adult-onset progressive muscular dystrophies. The LGMDs are divided into
type 1 (autosomal dominant) and type 2 (autosomal recessive) categories. Type 1 appears to account for
less than 10% of cases to date, although this may be due in part to a lower rate of diagnosis in these
milder autosomal dominant conditions.
Cardiac complications are relatively frequent in types 1B, 1D, 2G, and 2I, but this list will likely expand in
the future as the various subtypes are studied in greater detail. Individuals with LGMD generally have
normal intelligence, although LGMD2K is associated with mental retardation. The genes that cause
certain LGMDs, such as 1B, 1C, 2B, 2I, and 2K, have also been associated with congenital muscular
dystrophy, other myopathies, and even primarily nonmuscular disorders. Specific mutations in CAV3 and
DYSF are associated with multiple phenotypes, raising the possibility that other genetic factors may
modify the phenotype.
LGMD1A: Myotilinopathy
The age of onset in LGMD1A is early adulthood. Initially, the weakness is proximal, but it becomes more
generalized as the disease progresses. These patients additionally have a distinctly dysarthric pattern of
speech. CK levels are markedly elevated.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 11 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

LGMD1A is caused by mutations in TTID, which encodes myotilin, a protein that forms part of the Z-disk in
the sarcomere (Hauser et al, 2000; Salmikangas et al, 1999). It appears to play a structural role, although
details remain unclear. Mutations in TTID cause Z-disk abnormalities, which may appear histologically as
rod-shaped structures resembling nemaline rods.
LGMD1B: Laminopathy
LGMD1B is characterized by proximal muscle weakness and a high rate of cardiac complications. The
latter makes sense in light of the genetic overlap with Emery-Dreifuss muscular dystrophy.
In addition to LGMD1B, mutations in LMNA can also cause Emery-Dreifuss muscular dystrophy, isolated
cardiomyopathy, lipodystrophy, and Hutchinson-Gilford progeria (De Sandre-Giovannoli et al, 2003;
Eriksson et al, 2003). It is unclear how a single gene can cause such diverse phenotypes in different
individuals. The two protein products, lamin A and lamin C, are produced by alternative splicing and
localize to the nuclear lamina.
LGMD1C: Caveolinopathy
Children with LGMD1C typically present at 5 years with proximal muscle weakness, calf hypertrophy, and
a Gowers sign. There have been occasional reports of exercise intolerance. CK levels are significantly
elevated.
Mutations in CAV3, the gene encoding caveolin 3, are responsible for cases of LGMD1C (Minetti et al,
1998). Other diseases caused by mutations in the same gene include rippling muscle disease (Betz et al,
2001; Kubisch et al, 2003), distal myopathy, and hyper-CKemia (Carbone et al, 2000). A single mutation
in CAV3 has been shown to cause rippling muscle disease with distal myopathy in some individuals, and
rippling muscle disease with LGMD in others.
LGMD1D
Onset of LGMD1D is typically in early adulthood, sometimes in late adolescence. Mild proximal weakness
may occur, but the most prominent manifestations are cardiac, including dilated cardiomyopathy and
cardiac conduction defects. Sudden death has occurred. The cardiac symptoms may precede the
weakness. CK levels are mildly elevated.
The responsible gene has not been identified. The locus has been linked to a 3-cM region of 6q23
(Messina et al, 1997). Two nearby laminins have been excluded as causative genes, but it is possible that
a previously unidentified laminin may be hiding in that region.
LGMD1E
Patients with LGMD1E have a proximal pattern of muscle weakness.
The gene has not been identified, but linkage analysis has narrowed the region to a 9-cM stretch on 7q
(Speer et al, 1999). The odds ratio for this region is not optimal; the genetic origins of this disorder remain
largely obscure.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 12 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

LGMD1F
LGMD1F is also known as vocal cord and pharyngeal weakness with autosomal dominant distal myopathy
(VCPDM), with onset typically in middle adulthood. The presentation may include voice change, finger
extensor weakness, peroneal weakness, shoulder girdle weakness, and dysphagia. CK levels may be
mildly elevated. Clinically it is a distal myopathy, and histologically there is little evidence for inflammation,
so it does not fit the typical picture of a muscular dystrophy.
The genetic locus has been narrowed to a 12-cM interval on 5q31 (Feit et al, 1998).
LGMD2A: Calpainopathy
To date, LGMD2A appears to be the most common single form of LGMD. It presents with progressive,
predominantly proximal weakness. Cardiac or facial involvement has not been documented thus far. The
severity varies widely, with many patients remaining ambulatory into the fifth decade. The phenotype does
not correlate consistently with calpain 3 expression levels in muscle tissue.
Mutations in CAPN3, which encodes the protein calpain 3, are responsible for LGMD2A (Richard et al,
1995). Following the initial report, a number of other mutations have been identified in various populations
(Dincer et al, 1997; Kawai et al, 1998; Penisson-Besnier et al, 1998; Richard et al, 1997; Topaloglu et al,
1997).
Deficiency of the calpain 3 protein has been documented on immunohistochemical analysis of muscle
tissue obtained from patients with LGMD2A (Anderson et al, 1998; Spencer et al, 1997). Calpain 3 is a
muscle-specific calcium-activated proteolytic enzyme, the first primarily nonstructural protein associated
with LGMD (Ono et al, 1998). Calpain 3 cleaves filamin 2, but the link between this function and the
phenotype remains unclear (Guyon et al, 2003a). The enzyme degrades quickly, making it difficult to
study. In human muscle biopsy tissue and mouse models, calpain 3 deficiency is associated with
myonuclear apoptosis and abnormal expression of NF-B and its inhibitor IB. As in other muscular
dystrophies, disruption of the sarcolemma in the mouse model was observed via injection of Evans blue
dye, which is membrane-impermeable. The relation of either phenomenon to the disease process is
unclear.
LGMD2B: Dysferlinopathy
LGMD2B presents with a proximal pattern of weakness, typically in childhood or early adulthood. CK
levels are markedly elevated. This disorder is milder than other autosomal recessive LGMDs; only about
10% of patients lose ambulation.
Mutations in DYSF, which encodes dysferlin, cause LGMD2B as well as Miyoshi myopathy and distal
myopathy with tibial onset (Bashir et al, 1998; Liu et al, 1998). The gene is large, consisting of 55 exons
and a 6243-bp open reading frame. The protein product is expressed mainly in skeletal and cardiac
muscles. Individual missense mutations have been observed to cause both LGMD2B and Miyoshi
myopathy in different individuals within the same family, while a single base pair deletion has been
observed to cause both Miyoshi myopathy and distal myopathy with tibial onset in different kindreds.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 13 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

LGMD2C-F: The Sarcoglycanopathies


The four sarcoglycanopathies present with progressive proximal muscle weakness, typically in the first
decade. LGMD2C, LGMD2D, LGMD2E, and LGMD2F correspond to -sarcoglycanopathy,
-sarcoglycanopathy, -sarcoglycanopathy, and -sarcoglycanopathy, respectively. SGCG, SGCA,
SGCB, and SGCD encode -, -, -, and -sarcoglycans and are the genes responsible for LMD2C,
LGMD2D, LGMD2E, and LGMD2F, respectively. The sarcoglycans associate with each other to form a
sarcolemmal protein complex (see below). Among them, LGMD2C (-sarcoglycanopathy) and LGMD2D
(-sarcoglycanopathy) tend to have a milder phenotype than LGMD2E (-sarcoglycanopathy) and
LGMD2F (-sarcoglycanopathy), although there is significant overlap between the range of phenotypes
seen in these disorders (OBrien, Kunkel, 2001). Symptomatic cardiomyopathy occurs in a minority of
patients, but subclinical cardiac disease is relatively common (Fadic et al, 1996; Gnecchi-Ruscone et al,
1999; Melacini et al, 1999; Prelle et al, 1998). As noted below, specific mutations have been noted to give
rise to a diversity of phenotypes, suggesting that the activity of modifying genes may have an impact on
the pathogenesis of these diseases.
LGMD2C (-sarcoglycanopathy) is caused by mutations in SGCG (McNally et al, 1996a; McNally et al,
1996b; Noguchi et al, 1995). A single mutation has been associated with a range of severity of the
disease (McNally et al, 1996b). This disease was initially identified in Tunisian kindreds, but reports of
SGCG mutations have been documented in other populations as well (Bonnemann et al, 2002; Buzzini,
Macmilan, 2001; Ginjaar et al, 2000; Lasa et al, 1998; Piccolo et al, 1996; Takano et al, 2000).
LGMD2D (-sarcoglycanopathy) is caused by mutations in SGCA (Piccolo et al, 1995; Roberds et al,
1993a; Roberds et al, 1994). It appears to be the most common among the sarcoglycanopathies, with a
prevalence of 5 to 10% among individuals with muscular dystrophy who do not have a dystrophinopathy
(Duggan et al, 1996; Duggan et al, 1997a). Most mutations appear to be in the extracellular domain
(Carrie et al, 1997). There is often a good correlation between the severity of the mutation, the degree of
protein deficiency, and the clinical phenotype (Eymard et al, 1997), but this is not uniform (Angelini et al,
1998; Higuchi et al, 1997).
LGMD2E (-sarcoglycanopathy) is caused by mutations in SGCB (Bonnemann et al, 1995; Lim et al,
1995). Genotype-phenotype correlations have been inconsistent in some cases (Bonnemann et al, 1998).
LGMD2F (-sarcoglycanopathy) is caused by mutations in SGCD (Duggan et al, 1997b; Nigro et al,
1996a; Nigro et al, 1996b).
LGMD2G
The onset of LGMD2G is typically in late childhood or adolescence with proximal weakness. In some
cases there may also be a distal pattern of weakness that resembles the phenotype of Miyoshi myopathy.
Many affected individuals lose the ability to ambulate independently by the third or fourth decade. Cardiac
complications occur frequently.
LGMD2G is caused by mutations in TCAP, which encodes telethonin (Moreira et al, 2000). Telethonin
localizes to the Z-disk of the sarcomere and is expressed in skeletal and cardiac muscles. Its specific
function is unknown, but it interacts with titin.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 14 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

LGMD2H
Patients with LGMD2H have mild weakness that is slowly progressive.
The disease is caused by mutations in TRIM32, which encodes a protein with the same name that is an
E3 ubiquitin ligase expressed in skeletal and cardiac muscle (Frosk et al, 2002). It is possible that a
deficiency of the enzyme would cause the accumulation of its substrate, but histologic examination has
not revealed any unusual deposits in muscle fibers.
LGMD2I
There are two clinical forms of LGMD2I. In the severe form, children present in the first 2 years of life with
hypotonia, motor delays, and muscle hypertrophy (which may sometimes extend to macroglossia). The
course is similar to that of DMD, with frequent cardiac complications and loss of ambulation in early
adolescence. Some severe cases may resemble the phenotype of congenital muscular dystrophy 1C,
which is caused by mutations in the same gene.
The milder variant has been observed in a Tunisian family, with later onset and a slower progression.
Ambulation is preserved in nearly all cases. CK levels are markedly elevated in both forms.
Mutations in FKRP, which encodes fukutin-related protein, cause LGMD2I, as well as congenital muscular
dystrophy 1C (Brockington et al, 2001b). In contrast to the genotype-phenotype correlations with dysferlin,
different mutations in FKRP cause the two different phenotypes, suggesting that the mutations themselves
are primary determinants of the clinical manifestations. Patients with LGMD2I frequently have a
c.826CA, p.L276I mutation. In the more severe cases, secondary deficiencies of laminin-2 and
-dystroglycan may be observed on muscle tissue immunohistochemistry, western blots, or both.
LGMD2J
Patients with LGMD2J have proximal weakness with elevated CK levels.
LGMD2J is caused by mutations in TTN, which encodes titin (Haravuori et al, 2001). Titin is a large
sarcomeric protein that connects the M-line to the Z-disk. Immunohistochemistry of muscle tissue
obtained from affected individuals demonstrates a secondary deficiency of calpain 3.
LGMD2K
Individuals with LGMD2K tend to have relatively mild proximal muscle weakness with mild muscle
hypertrophy and marked elevations in serum CK levels. Onset in the small Turkish cohort identified so far
has been between 1 and 3 years, and patients appear to remain ambulatory at least until late adolescence
(Balci et al, 2005). Distinctive features include microcephaly and mental retardation, although no structural
abnormalities can be identified on brain imaging (Dincer et al, 2003). Muscle biopsies obtained from these
individuals demonstrated a reduction in -dystroglycan staining, suggesting the possibility of a primary
glycosylation defect. A mutation in POMT1 was identified (A200P), confirming that the primary
biochemical defect is in glycosylation (Balci et al, 2005).

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 15 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Clinical Presentation and Natural History: Congenital Muscular Dystrophies


The congenital muscular dystrophies (MDCs) are a growing category of degenerative muscle diseases
that are symptomatic from birth. These include some of the most severe and devastating muscular
dystrophies. There are three broad groups of MDCs: mild [laminin-2 (merosin)-positive and normal
cognition], moderate [laminin-2 (merosin)-negative and normal cognition], and severe
(laminin-2-negative with significant central nervous system abnormalities). Patients with the moderate
form have primary or secondary deficiencies in laminin-2, may have white matter abnormalities on
magnetic resonance imaging (MRI), and have normal or mildly delayed cognitive development. The
severe MDCs are accompanied by significant brain malformations and mental retardation, and typically
have a rapidly progressive course. Immunohistochemical staining typically shows a partial secondary
deficiency of laminin-2 in the severe forms.
Mild, Laminin-2 (Merosin)-Positive Congenital Muscular Dystrophies
The milder MDCs are characterized by normal staining of laminin-2 on immunohistochemical staining of
muscle tissue sections. Structural brain lesions are not typically found, and cognitive abilities are usually
normal. There are many cases of such MDCs that cannot be further characterized, but genetic mutations
have been linked to two MDC syndromes: Ullrich disease and rigid spine syndrome.
Infants with Ullrich disease have dramatic hypotonia, motor delays, and muscle atrophy. The diagnosis
may be suggested by the unusual combination of distal joint hyperextensibility and proximal joint
contractures. Cognition is normal. Some affected children walk for several years, but by the later stages
ambulation is always lost.
Mutations in COL6A1, COL6A2, and COL6A3 have been demonstrated to cause some cases of Ullrich
disease (Camacho Vanegas et al, 2001; Demir et al, 2002; Higuchi et al, 2001). These genes encode
collagen, type VI, 1, 2, and 3, respectively, and were initially associated with Bethlem myopathy
(Jobsis et al, 1996; Pan et al, 1998). Correspondingly, collagen VI has been found to be deficient on
immunohistochemistry of muscle tissue obtained from patients, whereas collagen IV expression is
upregulated, most likely as a secondary response. Collagen VI is expressed widely in connective tissues
and anchors collagen I/III fibrils to basement membranes.
Children with rigid spine syndrome generally have mild weakness. Most of them can eventually walk. CK
levels are normal or mildly elevated. However, they are at increased risk of developing respiratory
insufficiency requiring mechanical ventilation, perhaps due to selective weakness in the diaphragm.
Rigid spine syndrome is caused by mutations in SEPN1, which encodes selenoprotein N
(Moghadaszadeh et al, 2001). This protein incorporates selenium in the form of selenocysteine. The
function of selenoprotein N remains obscures, but selenium deficiency causes cardiomyopathy in humans
and muscular dystrophy in livestock.
Most children with laminin-2 (merosin)-positive congenital muscular dystrophy follow a mild course, aside
from the respiratory issues noted above. Further study may reveal other genes associated with this class
of diseases.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 16 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Moderate, Laminin-2 (Merosin)-Negative Congenital Muscular Dystrophy


The moderate forms of MDC are characterized by a more severe degree of motor involvement than the
mild forms with primary or secondary deficiencies in laminin-2. Neuroimaging typically reveals diffuse
white matter lesions in primary laminin-2 deficiency, but cognition is generally preserved.
MDC1A, or primary laminin-2 (merosin) deficiency, is the most common MDC, constituting about 40% of
all cases of MDC (Helbling-Leclerc et al, 1995; Tome et al, 1994). Infants present in the first 6 months of
life with muscle weakness, hypotonia, and contractures. Most affected individuals never walk
independently. Serum CK levels are mildly to moderately elevated. Diffuse white matter lesions are
evident on brain MRI, possibly due to merosin deficiency in the cerebral arteries (Villanova et al, 1996).
However, the gray matter and intelligence are generally preserved (Fig. 216-2). There have been some
reports of mental retardation. In some cases, seizures, cardiac complications, and neuronal migration
defects have been observed. The phenotype is sometimes milder (Sewry et al, 1997) and occasionally
presents as an inflammatory myopathy (Pegoraro et al, 1996). A milder variant resembling LGMD has
been described, although some of these patients may actually have LGMD2I.

Axial T2-weighted magnetic resonance imaging scan of the brain demonstrates marked hyperintense
signal in the cerebral white matter in a patient with merosin-deficient congenital muscular dystrophy.
Mutations in LAMA2 cause primary laminin-2 deficiency (Helbling-Leclerc et al, 1995; Hillaire et al, 1994;
Tome et al, 1994). In some cases, a missense mutation may cause a partial primary deficiency of
laminin-2 (Nissinen et al, 1996).

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 17 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Some MDCs with secondary laminin-2 deficiency also cause a moderately severe phenotype, with
sparing of the central nervous system. These include MDC1C with fukutin-related protein deficiency
(Brockington et al, 2001a).
Severe Congenital Muscular Dystrophies
In order of generally increasing phenotypic severity, the three classic severe MDCs are Fukuyama
congenital muscular dystrophy (FCMD), muscle-eye-brain disease (MEB), and Walker-Warburg syndrome
(WWS). They share a more severe course than the other MDCs, structural abnormalities in both the white
and gray matter of the brain, and significant mental retardation. The affected proteins all localize to the
Golgi apparatus and all appear to play a role in protein glycosylation, specifically of components of
-dystroglycan, which provides a biochemical link to laminin-2. Immunohistochemistry of muscle
sections obtained from affected patients demonstrates secondary deficiencies of both laminin-2 and
-dystroglycan. More recently, a mutation in LARGE, which encodes a glycosyltransferase with the same
name, has been linked to a form of MDC (MDC1D) with severe weakness, minimal ambulation, and
severe mental retardation (Longman et al, 2003).
FCMD is most common in Japan, with an incidence of about 1 in 10,000 live births (Fukuyama et al, 1981;
Kamoshita et al, 1976; Kobayashi et al, 1998b). Infants with FCMD have severe weakness and hypotonia.
They rarely walk independently. Brain lesions may include cortical and cerebellar dysgenesis (especially
polymicrogyria), cerebellar cysts, hydrocephalus, and white matter abnormalities, resulting in mental
retardation and epilepsy in many cases (Aida et al, 1996; Aida et al, 1994). There is a broad range of
clinical severity (Kondo-Iida et al, 1997; Saito et al, 2000; Yoshioka, Kuroki, 1994; Yoshioka et al, 1999).
Most affected individuals eventually sit but do not walk, and life expectancy may extend to early
adulthood. However, in the most severe cases, motor development is very poor with survival only for a
few years, and in rare cases at the milder end of the spectrum, patients may be able to walk.
Mutations in FCMD, which encode fukutin, cause FCMD. The most common mutation is a 3-kb
retrotransposal insertion in the 3 untranslated region of the gene (Kobayashi et al, 1998a). Point
mutations have also been reported. In either case, the protein is completely absent in muscle tissue.
Fukutin plays a role in glycosylation of -dystroglycan.
Infants with MEB have severe weakness with hypotonia; most do not become ambulatory. The ocular
problems include myopia, glaucoma, optic disc pallor, and retinal hypoplasia. Structural brain lesions
include lissencephaly, a flattened brainstem, cerebellar hypoplasia, and white matter changes. Mental
retardation is uniformly present, and epilepsy commonly occurs. With supportive care, life expectancy can
be into early adulthood.
Mutations in POMGnT1 (O-mannose -1,2-N-acetylglucosaminyltransferase), which encodes a protein
with the same name, cause MEB (Yoshida et al, 2001). The protein product is a glycosyltransferase found
in the Golgi apparatus that catalyzes the transfer of N-acetylglucosamine from UDP-GlcNAc to
O-mannosyl glycoproteins, which is the second step in the biosynthesis of O-mannosylglycan, a
component of -dystroglycan.
WWS is perhaps the most severe muscular dystrophy. Patients rarely survive infancy, and a number
probably die in utero, since mothers of affected infants often report a history of miscarriages. The clinical
presentation includes severe weakness and hypotonia. Brain malformations may include cobblestone
lissencephaly, agenesis of the corpus callosum, cerebellar hypoplasia, hydrocephalus, and extensive

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 18 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

white matter abnormalities. Other problems may include buphthalmos, glaucoma, other eye abnormalities,
and testicular defects. Serum CK levels are moderately elevated.
To date, the most common causative gene appears to be POMT1 (O-mannosyltransferase)
(Beltran-Valero de Bernabe et al, 2002). Mutations in this gene have been identified in six different
families. Isolated cases of WWS have been associated with mutations in FCMD and FKRP.
Clinical Presentation and Natural History: Distal Myopathies with Dystrophic Features
The distal myopathies have in recent years expanded to become a sizable class of disorders. Two of
them, Miyoshi myopathy and tibial muscular dystrophy (Udd myopathy), are allelic with limb girdle
muscular dystrophies 2B and 2J, respectively, and have dystrophic findings on muscle biopsy. These two
will be described briefly. The other classic distal myopathies, Welander myopathy, Nonaka myopathy, and
Laing myopathy, are beyond the scope of this discussion.
Miyoshi Myopathy
Miyoshi myopathy was recognized and described separately in Japan (Miyoshi et al, 1977) and the United
States (Markesbery et al, 1977). Inheritance is autosomal recessive, and onset is typically in late
adolescence or early adulthood, with early involvement of the gastrocnemius muscle. A cardinal sign is
difficulty or inability to stand on tiptoes. Other skeletal muscles may also be affected, but to a lesser
extent. CK elevation is typical, as are myopathic findings on electromyography. Muscle histology
demonstrates a dystrophic pattern (Miyoshi et al, 1986), with advanced fibrosis in gastrocnemius,
intermediate findings in biceps femoris, and only scattered necrotic fibers in vastus lateralis, suggesting
that biceps femoris is the optimal muscle to sample for analysis (Barohn et al, 1991). Miyoshi myopathy is
caused by homozygous or compound heterozygous mutations in DYSF, the same gene that causes
LGMD2B (Liu et al, 1998).
Tibial Muscular Dystrophy
Tibial muscular dystrophy (Udd myopathy) is inherited in an autosomal dominant manner, with onset later
than 35 years and selective involvement of the tibialis anterior muscle. Extensor digitorum brevis, which is
also innervated by the peroneal nerve, is preserved. Serum CK levels are mildly elevated in the majority of
patients, electromyography of affected muscles demonstrates myopathic features, and dystrophic findings
are present on muscle biopsy, sometimes accompanied by vacuolar changes (Udd et al, 1993). The
selection of the muscle to be biopsied is important, as asymptomatic muscles may show few histologic
signs of disease (Udd et al, 1993). Tibial muscular dystrophy is caused by mutations in TTN, the same
gene implicated in LGMD2J (Hackman et al, 2002).
In rare cases, a homozygous mutation in DYSF may cause a similar phenotype of distal myopathy with
anterior tibial onset (DMAT) (OMIM 606768), with an autosomal recessive inheritance (Liu et al, 1998).
Clinical Presentation and Natural History: Syndromic and Miscellaneous Muscular Dystrophies
All of the muscular dystrophies in this section have fairly unique phenotypes and cannot easily be included
in any of the above categories.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 19 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Bethlem Myopathy
The initial description of Bethlem myopathy outlined a relatively benign autosomal dominant disorder
characterized by onset around age 5 years, with contractures at the elbows and interphalangeal joints
(Bethlem, Wijngaarden, 1976). This classic presentation has generally been confirmed by other
observers, with a broader range of severity than was first recognized. On the milder end of the spectrum,
symptoms may be very subtle in relatives of affected individuals, and onset may be delayed until middle
adulthood (Merlini et al, 1994). Longer follow-up and evaluation of larger cohorts have demonstrated that
affected children may have congenital hypotonia and contractures, and that after the age of 50 years, the
majority of patients require the use of a wheelchair (Jobsis et al, 1999).
Mutations in COL6A1, COL6A2, and COL6A3 cause Bethlem myopathy as well as Ullrich congenital
muscular dystrophy (Jobsis et al, 1996; Pan et al, 1998).
Emery-Dreifuss Muscular Dystrophy
The symptoms of Emery-Dreifuss muscular dystrophy (EDMD) begin in the first decade of life, and may
including toe-walking and limitation of movement at the elbows, neck, and spine. The selective pattern of
contractures in EDMD is striking and unique, involving the posterior neck, elbows, and heel cords. In early
childhood, only the heel cord contractures may be present. Contractures are typically more prominent
than muscle weakness. Muscle weakness and atrophy in a humeroperoneal distribution typically begin in
the second decade. Cardiac conduction defects leading to heart block are common in affected individuals
and some carriers and may be life-threatening, especially during anesthesia (Hopkins et al, 1981; Jensen,
1996). This complication may be treated by the insertion of a cardiac pacemaker. Intelligence is usually
normal. A clinical variant resembling LGMD has been identified (Muntoni et al, 1998).
Two genes are known to cause EDMD: EMD and LMNA. EMD is a 2-kb, 6-exon gene located at Xq28
(Bione et al, 1994). After the gene was identified, further mutations were found (Bione et al, 1995;
Ichikawa et al, 1997; Klauck et al, 1995; Nigro et al, 1995; Wulff et al, 1997a; Wulff et al, 1997b; Yamada,
Kobayashi, 1996). Large inverted repeats flank the EMD gene, making it susceptible to large-scale partial
or whole-gene deletions (Small et al, 1997; Small, Warren, 1998). LMNA is the second causative gene
and is located at 1q21.2-q21.3 (Bonne et al, 1999). The disease is inherited in an X-linked recessive
fashion when associated with EMD and in an autosomal dominant or recessive fashion when LMNA is
involved (Raffaele Di Barletta et al, 2000). Mutations in LMNA can cause several other diseases, including
LGMD1B, cardiomyopathy, lipodystrophy, and Hutchinson-Gilford progeria.
Emerin, the protein product of EMD, is expressed ubiquitously but is predominantly found in skeletal
muscle, heart, colon, testis, ovary, and pancreas. Emerin localizes to the inner nuclear membrane via the
hydrophobic C-terminal domain (Manilal et al, 1996; Nagano et al, 1996). Emerin and lamin A interact at
the nuclear lamina (Clements et al, 2000; Manilal et al, 1998).
Facioscapulohumeral Muscular Dystrophy
The onset of facioscapulohumeral muscular dystrophy (FSHD) is typically in the second decade. Males
are more severely affected than females (Zatz et al, 1998). Affected individuals have a distinct pattern of
muscle wasting and weakness. The muscles most characteristically affected are in the face, shoulder
girdle, and upper arm. Pelvic girdle, forearm, and peroneal muscles may also be affected later.
Sensorineural hearing loss and retinal vasculopathy (telangiectasias) may occur, requiring periodic
hearing evaluations and ophthalmologic monitoring (Funakoshi et al, 1998). Intelligence is often normal
but a subset of patients develop mental retardation (Funakoshi et al, 1998). The majority of patients

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 20 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

remain ambulatory. Cardiac involvement is variable (Laforet et al, 1998). Phenotypic variants that have
been linked to the same genetic locus include scapuloperoneal and scapulohumeral dystrophies.
The exact genetic etiology of FSHD has been a longstanding mystery, but new clues may be emerging.
The disease has been mapped to a region on 4q35 (Upadhyaya et al, 1992; Wijmenga et al, 1990;
Wijmenga et al, 1991), and patients with FSHD have deletions in a D4Z4 tandem repeat sequence (Hewitt
et al, 1994; van Deutekom et al, 1993), making it possible to perform genetic testing (Bakker et al, 1996).
Each D4Z4 sequence is 3.3 kb in length and contains two homeobox domains. Affected individuals have
fewer than 11 of these repeats. The severity of the deletion tends to correlate with the severity of the
disease (Lunt et al, 1995). Many cases are familial, while sporadic cases may arise from germline
mosaicism (Roques et al, 1998). The deletion is stably inherited within families. Other factors appear to be
involved in the pathogenesis, as monozygotic twins with identical D4Z4 deletions have been reported to
have divergent phenotypes (Tupler et al, 1998). Also complicating matters is the presence of a
homologous region on 10q that is not pathogenic (Cacurri et al, 1998); a method was developed to
distinguish between the two (Deidda et al, 1996; Upadhyaya et al, 1997).
The mystery has surrounded the lack of a known coding region within the D4Z4 sequence. Neither an
RNA transcript nor a protein product has been identified. The most likely mechanism appears to be
position effect variegation. This was suggested by the upregulation of a number of genes upstream of the
D4Z4 region. Ordinarily, a protein complex consisting of YY1, HMGB2, and nucleolin binds to the D4Z4
region and mediates transcriptional repression of the upstream genes, including ANT1, FRG1, and FRG2.
A deletion in the D4Z4 region could conceivably disrupt this balance, leading to overexpression of the
upstream genes (Gabellini et al, 2002). Overexpression of ANT1, FRG1, and FRG2 in a mouse model
resulted in a dystrophic phenotype in the mice who overexpressed FRG1 but not the other two genes; this
was demonstrated via observation of kyphosis, muscle atrophy, histology, and alternative splicing patterns
of Tnnt3 and Mtmr1 (Gabellini et al, 2006). The alternative splicing of TNNT3 and MTMR1 was also
shown in muscle cell cultures derived from human FSHD patients (Gabellini et al, 2006). These results
suggest that upregulation of FRG1 is the mechanism by which FSHD develops, but further studies may be
required to confirm this.
Oculopharyngeal muscular dystrophy
Oculopharyngeal muscular dystrophy (OPMD) was originally described as an inherited syndrome
characterized by ptosis and dysphagia (Taylor, 1915). In many regions it is rare, with a prevalence of 1 in
200,000 for the autosomal dominant type in France (Brunet et al, 1990). However, certain populations
have a higher prevalence, including French-Canadians (Brais et al, 1997) and Bukhara Jews living in
Israel (Blumen et al, 1997). The presentation of the dominant form of OPMD typically occurs between
ages 50 and 70 with dysphagia, ptosis, and proximal muscle weakness (Victor et al, 1962). Other signs
may include facial weakness, limitation of upgaze, dysphonia, and tongue atrophy and weakness
(Bouchard et al, 1997). The recessive phenotype is similar, although the severity may be milder (Fried et
al, 1975). Pathologically, nuclear filamentous inclusions are found in muscle fibers of affected individuals
with both dominant and recessive forms of the disease and are regarded to be specific histologic markers
for the disease (Blumen et al, 1996; Brais et al, 1998; Coquet et al, 1990; Tome, Fardeau, 1980).
OPMD is the only muscular dystrophy caused by a triplet repeat expansion, unless myotonic dystrophy is
regarded primarily as a muscular dystrophy rather than a myotonic disorder. In normal individuals,
PABP2, which encodes poly(A) binding protein 2, contains a (GCG) 6 repeat in exon 1. Individuals
affected with OPMD have a (GCG) 8-13 expansion in exon 1 (Brais et al, 1998). In contrast to other triplet
repeat diseases, the expansion has a low copy number and is stably inherited. The disease is typically

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 21 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

inherited in an autosomal dominant fashion, but a borderline (GCG) 7 expansion on the other allele may
worsen the phenotype typically expected for the size of the longer expansion. In rare cases, a
homozygous (GCG) 7 expansion can cause a mild form of OPMD that is inherited in an autosomal
recessive manner.
Genetics and Biochemistry
Dystrophin
The complete sequence of the DMD gene was published soon after the initial exons were cloned (Koenig
et al, 1987). It is the largest gene identified to date with respect to the total number of base pairs (2.3
million) ( http://genome.ucsc.edu), although TTN (see below) has a greater number of exons and a larger
protein product (titin). The DMD gene has 79 exons (Coffey et al, 1992; Roberts et al, 1993) and is
transcribed at about 40 nucleotides per second, with a total transcription time of 16 hours (Tennyson et al,
1995). This was the first description of cotranscriptional splicing in a mammalian cell (Tennyson et al,
1995). Deletions form approximately two-thirds of mutations found in affected patients, and they tend to
cluster in exons 2 to 20 and 44 to 53 (Gillard et al, 1989; Koenig et al, 1989). The intronic sequences
between these exons tend to be unusually long, nearly 200 kb, which accounts for their vulnerability to
deletions.
In skeletal muscle, DMD encodes a 427-kDa protein called dystrophin that represents 0.002% of total
muscle protein (Hoffman et al, 1987) and 5% of membrane cytoskeleton (Ohlendieck, Campbell, 1991). It
is a monomeric protein (Chan, Kunkel, 1997; Rybakova, Ervasti, 1997) that was first localized to the
sarcolemmal region (Arahata et al, 1988; Bonilla et al, 1988; Zubrzycka-Gaarn et al, 1988), and then more
specifically to the subsarcolemma (Watkins et al, 1988). The skeletal muscle isoform is composed of 3685
amino acids and has four major domains: amino terminus, rod domain, cysteine-rich domain, and
carboxy-terminus (Koenig et al, 1988). The amino terminus binds to F-actin and resembles the
corresponding regions of -actinin and -spectrin (Rybakova et al, 1996). The rod-like domain contains 24
spectrin-like repeats and 4 hinge segments, and is 15 to 20 nm from the cytoplasmic face of the
sarcolemma (Cullen et al, 1991; Koenig, Kunkel, 1990). Hinge 4 and the cysteine-rich domain of
dystrophin bind to the 15 C-terminal amino acids of -dystroglycan, thus connecting dystrophin to the
extracellular matrix (Jung et al, 1995; Koenig et al, 1988). Both the amino terminus and rod domains bind
to F-actin (Rybakova et al, 1996); in mice, a deletion of just the amino terminus results in a mild
phenotype (Corrado et al, 1994; Corrado et al, 1996), while deletions in both domains cause a more
severe phenotype (Cox et al, 1994), suggesting that the F-actin binding property is an important function
of dystrophin.
There is also a high concentration of dystrophin at the myotendinous junction (Byers et al, 1991) and the
postsynaptic portion of the neuromuscular junction (Huard et al, 1992b). It appears that dystrophin has a
structural function and protects muscle from damage during contractions (Petrof et al, 1993; Weller et al,
1990), although it may also play a more dynamic role such as regulation of the repair and perfusion of
muscle through its association with syntrophin and in turn nitric oxide synthase (see below) (Brenman et
al, 1996; Rando, 2001). Dystrophin is also present in the subplasmalemmal region of cardiac and smooth
muscle; its expression in the latter is relatively patchy (Byers et al, 1991; Hoffman et al, 1988b).
There are three major sources of variation in dystrophin expression that lead to the various known
isoforms: alternative promoters, variable lengths of transcripts, and splice variations in the carboxy
terminus. Full-length transcripts that differ by only a few base pairs are produced in muscle, cardiac
Purkinje fibers, and brain with alternative promoters (Bies et al, 1992; Boyce et al, 1991; Chamberlain et

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 22 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

al, 1988b; Nudel et al, 1988). The brain also expresses a shorter, Dp140 isoform postsynaptically (Lidov
et al, 1993; Lidov et al, 1990; Lidov et al, 1995). The Dp116 isoform is present in Schwann cells (Byers et
al, 1993), Dp260 in retina (DSouza et al, 1995), and Dp71 in other nonmuscle tissues, including a
different location in the retina (Howard et al, 1998; Rapaport et al, 1992; Yaffe et al, 1992). Further
diversity in dystrophin expression is created by splice variations at the carboxy terminus (Feener et al,
1989; Lidov, 1996).
Deficiency of dystrophin appears to cause mechanical weakness in the sarcolemma, followed by
disruptions of the sarcolemma during muscle contraction (Menke, Jockusch, 1991). This leads to an influx
of calcium (Carpenter, Karpati, 1989) into the muscle fiber, followed by mitochondrial calcium overload
that leads to impairment of oxidative phosphorylation and muscle fiber necrosis (Mezon et al, 1974;
Wrogemann et al, 1970; Wrogemann et al, 1973).
Dystrophin-Associated Protein Complex
Several years after the cloning of dystrophin, it became apparent that it was associated with a number of
other proteins, including a sarcolemmal glycoprotein (Campbell, Kahl, 1989; Ervasti et al, 1990; Yoshida,
Ozawa, 1990). This large cluster of proteins forms the DAPC (Fig. 216-3) (Ervasti, Campbell, 1991). The
complex has traditionally been divided into three clusters (Dalkilic, Kunkel, 2003; Yoshida, Ozawa, 1990),
but the recent discovery that biglycan links the sarcoglycan and dystroglycan clusters suggests that the
complex may alternatively be divided into two main categories: sarcolemmal and subsarcolemmal.

The current knowledge of the cellular localization of the dystrophin-associated protein complex, as well as
many of the other proteins involved directly or indirectly in the pathogenesis of the muscular dystrophies.
The proteins located at the sarcolemma include -dystroglycan and -dystroglycan, biglycan,
-sarcoglycan (Piccolo et al, 1995; Roberds et al, 1993a), -sarcoglycan (Bonnemann et al, 1995;
Bonnemann et al, 1996; Lim et al, 1995), -sarcoglycan (McNally et al, 1996a; McNally et al, 1996b;

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 23 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Noguchi et al, 1995), -sarcoglycan (Jung et al, 1996; Nigro et al, 1996a; Nigro et al, 1996b),
e-sarcoglycan (Ettinger et al, 1997; McNally et al, 1998), and sarcospan (Crosbie et al, 1997; Crosbie et al,
1999). Dystrophin binds to -dystroglycan (43 kDa), which binds to -dystroglycan (156 kDa), which in
turn binds to laminin-2, part of the extracellular matrix (Ibraghimov-Beskrovnaya et al, 1992). A single
gene, DAG1, encodes both - and -dystroglycan, which contribute to basement membrane assembly
(Henry, Campbell, 1998). Based on embryonic lethality in mouse models and the lack of an identified
human disease resulting from dystroglycan deficiency, it is most likely that primary dystroglycan deficiency
is an embryonic lethal mutation in humans (Williamson et al, 1997). -dystroglycans association with
Grb2 (Oak et al, 2001) suggests that it plays a signaling as well as structural role at the sarcolemma
(Yang et al, 1995). The sarcoglycans are very similar to one another. Each one has a single
transmembrane domain, small intracellular domain, and large extracellular domain. They range from 35 to
50 kDa, and all are glycosylated.
Biglycan is a proteoglycan that associates with -dystroglycan, - and -sarcoglycan, and collagen VI,
linking the dystroglycan cluster to the sarcoglycan cluster in the sarcolemma (Bowe et al, 2000; Wiberg et
al, 2001; Wiberg et al, 2002). The link to collagen VI also provides a second link to the extracellular matrix
for -dystroglycan. -, -, -, and -sarcoglycan are linked with LGMD2C-F (see above). However,
deficiencies in -sarcoglycan cause myoclonus-dystonia syndrome rather than a muscular dystrophy
(Asmus et al, 2002; Zimprich et al, 2001). Maternal imprinting plays a role in the pathogenesis of this
disorder. -sarcoglycan replaces -sarcoglycan in smooth muscle (Straub et al, 1999). Sarcospan
interacts with the sarcoglycan complex (Crosbie et al, 1999). A small portion of the filamin C (gene symbol
FLNC, gene previously known as FLN2 and protein previously known as filamin) in the muscle fiber
interacts with - and -sarcoglycan, and its localization is altered in DMD and LGMD (Thompson et al,
2000). Calpain 3 associates with and cleaves filamin C (Guyon et al, 2003a). Most of the filamin C
localizes to the sarcomeric Z disk (see below).
The subsarcolemmal component of the DAPC includes dystrophin itself, 1-, 1-, and 2-syntrophin
(Adams et al, 1993; Ahn et al, 1996; Ahn et al, 1994), - and -dystrobrevin (Peters et al, 1997; Puca et
al, 1998; Sadoulet-Puccio et al, 1996), and nitric oxide synthase (Crawford et al, 2000; Grady et al, 1999).
The carboxy terminus coiled coil domains of dystrophin and the dystrobrevins associate (Sadoulet-Puccio
et al, 1997). Syntrophins bind to both dystrophin and the dystrobrevins (Ahn, Kunkel, 1995; Suzuki et al,
1995). Nitric oxide synthase binds to the syntrophins (Hillier et al, 1999), is absent in DMD and BMD
(Brenman et al, 1995; Chao et al, 1996), and is displaced in the dystrophic mouse model of -dystrobrevin
deficiency, impairing nitric oxide-mediated signaling (Grady et al, 1999). The -syntrophin null mouse
does not have a dystrophic phenotype, but the muscle tissue lacks nitric oxide synthase and has other
mild histologic abnormalities (Adams et al, 2000). These findings suggest that the DAPC potentially plays
a role in signaling, muscle fiber repair, or both in addition to its structural function (Brenman et al, 1996;
Rando, 2001). Curiously, -dystrobrevin null mice do not have perceptible phenotypic abnormalities
(Grady et al, 2006). nNOS and eNOS null mice have signaling abnormalities in skeletal muscle (Lau et al,
2000). F-actin binds to the amino terminus of dystrophin (Levine et al, 1992), and laminin-2 binds to
-dystroglycan, but these are generally not considered part of the DAPC proper.
When other proteins in the DAPC are deficient, as is found in other muscular dystrophies, the effect on
dystrophin expression is variable. In some cases, dystrophin expression is normal (Ben Jelloun-Dellagi et
al, 1990).

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 24 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Other Sarcolemmal Proteins


Utrophin is a close analogue of dystrophin (Buckle et al, 1990; Ishiura et al, 1990; Khurana et al, 1990;
Love et al, 1989; Nicholson et al, 1989) that is expressed throughout the sarcolemma during development
but localizes only to the neuromuscular and myotendinous junctions in mature muscle (Karpati et al,
1993b; Khurana et al, 1991; Ohlendieck et al, 1991; Voit et al, 1991). In mice, utrophin deficiency alone
produces only mild abnormalities (Deconinck AE et al, 1997a; Grady et al, 1997a), while utrophin
deficiency in mdx mice worsens the mdx phenotype (Deconinck AE et al, 1997b; Grady et al, 1997b), and
utrophin overexpression improves the mdx phenotype (Deconinck N et al, 1997; Rafael et al, 1998). This
suggests that utrophin may be able to compensate to some extent for dystrophin deficiency.
Caveolae are 50- to 100-nm invaginations of the plasma membrane of most cells, including the
sarcolemma of muscle fibers. Sarcolemmal caveolae contain dysferlin and caveolin 3, which interact with
each other (Matsuda et al, 2001; Tang et al, 1996). Caveolins participate in membrane trafficking, sorting,
transport, and signal transduction. Caveolin 3 is specific to muscle, interacts with -dystroglycan, and
colocalizes with dystrophin, linking it to the DAPC (Song et al, 1996; Sotgia et al, 2000). It forms a hairpin
loop within the sarcolemma, enabling both the amino and carboxy terminals to come in contact with the
cytoplasm. Abnormal expression of caveolin 3 in mouse models leads to a deficiency of caveolae and
abnormalities in the T tubule system, while in humans it causes LGMD. Dystrophin deficiency leads to
overexpression of caveolin 3 (Repetto et al, 1999), while overexpression of caveolin 3 leads to dystrophin
deficiency (Galbiati et al, 2000).
Dysferlin is homologous to the Caenorhabditis elegans protein fer-1, which plays a role in vesicle fusion
(Achanzar, Ward, 1997). In human tissue, dysferlin has been demonstrated to participate in the
calcium-dependent process of membrane resealing after sarcolemmal injury, suggesting that this repair
mechanism is defective in at least some forms of muscular dystrophy (Bansal, 2003; Lennon et al, 2003).
Dysferlin deficiency can cause several different muscle disorders (see LGMD2B and Miyoshi myopathy
above), suggesting that modifying genes influence the phenotype (Liu et al, 1998; Weiler et al, 1999).
Integrin 7 and integrin 1 form a small laminin receptor complex in the sarcolemma with no apparent
direct links to the DAPC. Integrin 7 deficiency is associated with a dystrophic phenotype in mice (Mayer
et al, 1997), but in humans it produces a form of congenital myopathy (Hayashi et al, 1998). Among other
functions, the complex appears to assist in the adhesion of muscle fibers to each other to generate
contractile force (Burkin, Kaufman, 1999).
Extracellular Matrix
There are two major types of structural proteins in the extracellular matrix (ECM) that have been linked to
muscular dystrophy: the laminins and collagens. LAMA2 is a 64-exon gene that encodes laminin-2,
which is deficient in MDC1A, merosin-negative congenital muscular dystrophy (Tome et al, 1994). Merosin
is the term for a laminin that contains an 2 heavy chain.
Sarcomere
Actin and myosin were the first major components of the sarcomere that were identified; they are major
components of the contractile apparatus. The structure of the sarcomere has become much more detailed
in recent years, and its complexity is astounding. Several sarcomeric proteins, including myotilin,
telethonin, and titin, have been implicated in various LGMDs. In the Z disk, titin interacts with telethonin
(Mues et al, 1998) and myotilin interacts with filamin C, -actinin, and F-actin (Salmikangas et al, 2003;
van der Ven et al, 2000). Deficiencies in titin cause secondary deficiencies in calpain 3 (Haravuori et al,

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 25 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

2001).
Golgi Complex
Several proteins that localize primarily in the Golgi complex play a role in glycosylation of the
dystroglycans that link the DAPC to the ECM. These proteins include fukutin, fukutin-related protein,
POMGnT1, POMT1, and most recently LARGE (Brockington et al, 2005; Michele et al, 2002). Deficiencies
in these proteins usually cause the severe forms of congenital muscular dystrophy, and occasionally
milder LGMDs. The connection to the dystroglycans and indirectly to laminin-2 provides a link between
some of the proteins that are responsible for the MDCs.
The POMT1 protein, like fukutin and POMGnT1, plays a role in glycosylation, as demonstrated by the
absence of -dystroglycan glycosylation in muscle sections obtained from affected individuals. The overall
amount of -dystroglycan is also reduced in muscle tissue from these patients. POMT1 is ubiquitous, but
peak levels of expression can be found in skeletal and cardiac muscle, fetal brain, and adult testis,
correlating with the phenotypic involvement.
The first glycosyltransferase associated with a muscular dystrophy was in the myd mouse, subsequently
renamed LARGE myd (Grewal et al, 2001; Longman et al, 2003). This mouse was originally thought to be a
model for FSHD but was subsequently found to have a mutation in Large, with 98% homology to human
Large (Grewal et al, 2001).
Sarcoplasmic Reticulum
The major protein in the sarcoplasmic reticulum associated with a muscular dystrophy is selenoprotein N,
1. A deficiency in this protein can cause rigid spine syndrome or multiminicore myopathy. The latter is,
strictly speaking, a variant of a congenital myopathy rather than a muscular dystrophy.
Nucleus
Several nuclear proteins, including lamin A/C, emerin, and poly(A) binding protein 2, are associated with
unique forms of muscular dystrophy. Lamin A/C is particularly intriguing as deficiencies may be
responsible for one of a broad range of diseases: Emery-Dreifuss muscular dystrophy, LGMD,
Charcot-Marie-Tooth disease type 2, Hutchinson-Gilford progeria (De Sandre-Giovannoli et al, 2003;
Eriksson et al, 2003), familial partial lipodystrophy, dilated cardiomyopathy, and mandibuloacral dysplasia.
Lamin A and lamin C are structural proteins of the nuclear envelope encoded by the same gene, LMNA,
via alternative splicing. Lamin A is 664 amino acids long, while lamin C is 572 amino acids in length.
Poly(A) binding protein 2 localizes to the nucleus (Krause et al, 1994) and is involved in the second phase
of mRNA polyadenylation (Wahle, 1991). A pathologic expansion of the GCG triplet repeat sequence
results in a longer polyalanine region that forms -sheet structures resistant to protease degradation
(Forood et al, 1995). Nuclear function may then be irreparably impaired.
FRG1 may be the gene that causes FSHD; its expression is increased indirectly via deletion of the D4Z4
region on 4q35 and plays a role in mRNA splicing (Gabellini et al, 2006).

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 26 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Enzymes
There are two enzymes linked to the LGMDs: calpain 3 and TRIM32. Calpain 3 is a calcium-activated
neutral protease while TRIM32 appears to be a ligase (Frosk et al, 2002; Richard et al, 1995).
Animal Models
The DAPC has been identified in a number of different organisms, among which several animal models of
muscular dystrophy have been identified. The animal models used most extensively are in the mouse,
dog, and zebrafish.
Mice and Other Rodents
The most extensively studied mouse model of DMD is the mdx mouse, a naturally occurring animal model
that has a nonsense mutation in exon 23 of mouse dystrophin (Bulfield et al, 1984; Ryder-Cook et al,
1988; Sicinski et al, 1989). Curiously, the phenotype of the mdx mouse is much milder than DMD in
humans (Durbeej, Campbell, 2002; Sicinski et al, 1989). Between 2 and 6 weeks of age, mdx hindlimb
muscles exhibit intense necrosis and regeneration, after which the muscles undergo stabilization, and
then hypertrophy and dystrophic degeneration as the mice age (Hoffman, Gorospe, 1991; McArdle et al,
1994). The one mdx mouse muscle that undergoes extensive degeneration similar to that found in human
DMD muscle is diaphragm (Cullen, Jaros, 1988; Stedman et al, 1991). A combined dystrophin-utrophin
mouse has been developed, and this model more closely replicates the phenotype of DMD (see
discussion of utrophin in Other Sarcolemmal Proteins, above).
Other mouse models of dystrophin deficiency have been developed. One that is becoming more widely
used in studies of muscular dystrophy is the mdx 5cv mouse, which has a mutation in exon 10 of
dystrophin generated by ethylnitrosurea mutagenesis (Chapman et al, 1989). This mutation creates a
novel splice site that induces aberrant splicing and an absence of dystrophin. The mdx 5cv phenotype is
more severe than the mdx one, but is still less severe than DMD. Mdx 5cv mouse muscles have 10 times
fewer revertant (dystrophin-positive) fibers than do those of mdx mice.
The differences in phenotypic severity among DMD and its various mouse models suggest that secondary
factors modulate the pathogenesis of the disease. This is also supported by the differential response of
specific muscle groups in mice and humans to dystrophin deficiency (Deconinck et al, 1998; Kaminski et
al, 1992; Karpati, Carpenter, 1986; Khurana et al, 1995; Matsumura et al, 1992; Moens et al, 1993).
Molecular differences between a limited number of muscle groups have been examined in mdx mice using
gene expression technology (Porter et al, 2002; Porter et al, 2003; Rouger et al, 2002; Tkatchenko et al,
2000; Tseng et al, 2002). One study in particular focused on a comparison of diaphragm and hindlimb
muscles (Porter et al, 2004), while another performed a broad survey of six different muscles: diaphragm,
soleus, extensor digitorum longus, quadriceps, gastrocnemius, and tibialis anterior (Haslett et al, 2005).
Among these six mouse muscles, soleus bears the closest molecular similarity to several human skeletal
muscles (Kho et al, 2006).
The other classic mouse model of muscular dystrophy is the naturally occurring dy/dy mouse, which has
been studied for decades (Meier, Southard, 1970). It has a more severe phenotype than the mdx mouse.
However, the genetic defect was mapped to the same locus as LAMA (laminin-2) rather than DMD, and
the mice were found to be deficient in the expression of laminin-2 (merosin) (Sunada et al, 1994; Xu et
al, 1994a). The genetic defect is a splice-site alteration that removes 57 amino acids from domain VI of
the protein, which would most likely disrupt self-aggregation of laminin heterotrimers (Sunada et al, 1995;
Xu et al, 1994b).

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 27 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

More recently, several mouse models of the sarcoglycanopathies have been developed. Cardiomyopathy
is a prominent feature of several of these strains, perhaps to a greater degree than is observed in human
sarcoglycanopathies. In the Sgca-null mouse, exons 2 and 3 and flanking introns of Sgca (the mouse
homologue of SGC, -sarcoglycan) were replaced by the neomycin resistance gene, creating a model of
LGMD2D (Duclos et al, 1998). The Sgca-null mouse has a phenotype of progressive muscular dystrophy
with histologic evidence of muscle necrosis and complete loss of the entire sarcoglycan complex and
sarcospan.
Via homologous recombination, exon 2 of murine -sarcoglycan was replaced by phosphoglycerate kinase
promoter/neomycin phosphotransferase cDNA (Hack et al, 1998). The resulting null mice displayed a
dystrophic myopathy and died prematurely of cardiomyopathy. There was complete loss of -sarcoglycan,
with secondary reductions in - and -sarcoglycan expression on immunohistochemistry.
Cardiomyopathy is also a hallmark of -sarcoglycan deficiency in mice and hamsters. Through
homologous recombination, exon 2 of -sarcoglycan was replaced by the neomycin resistance gene to
create Sgcd-null mice (Coral-Vazquez et al, 1999). Extensive pathologic disruptions were observed in the
skeletal muscles of these mice at an early age, as well as severe cardiomyopathy and histologic
abnormalities in the smooth muscle of the coronary vasculature. The naturally occurring BIO14.6 Syrian
hamster has a homozygous deletion in -sarcoglycan and develops a significant fatal cardiomyopathy but
only mild muscular dystrophy (Nigro et al, 1997; Roberds et al, 1993b; Sakamoto et al, 1997).
The naturally occurring myd mouse has an abnormal gait and dystrophic muscle histology. It was found to
have a frameshift deletion of exons 4-7 of the Large gene, which encodes a glycosyltransferase (Grewal
et al, 2001). Accordingly, the skeletal muscle has a secondary deficiency of -dystroglycan, while the
sarcoglycans and -dystroglycan remain intact.
A biglycan null mouse was recently generated by disrupting exon 2 of the bgn gene. Skeletal muscles
from these mice exhibit mild histologic abnormalities with secondary changes in the DAPC (Mercado et al,
2006).
Dogs
The xmd golden retriever is a naturally occurring dog model of DMD with absent RNA and protein
expression of dystrophin (Cooper et al, 1988). The genetic lesion is a point mutation that causes exon
skipping (Sharp et al, 1992). It more closely replicates the human phenotype than the currently available
mouse models, with marked elevations of CK, dystrophic findings on muscle histology, and clinical
weakness (gait difficulty) developing by age 8 to 10 weeks (Cooper et al, 1988; Kornegay et al, 1988;
Valentine et al, 1990). The xmd dog is also much closer to humans in size compared to the mdx mouse,
but is not as widely used in research studies due to the longer reproductive cycle and higher costs
associated with research involving these animals.
Zebrafish
In recent years, increasingly detailed investigations have been conducted of zebrafish (Danio rerio ) as
potential animal models for a number of diseases, including the muscular dystrophies. They have several
properties that distinguish them from mammalian models and contribute to their utility in the study of
muscle disease: short gestational periods, large litter sizes, and translucent bodies. Most components of
the DAPC have orthologues in zebrafish, and expression of the sarcoglycans, dystroglycan, and
dystrophin has been confirmed by immunoblotting (Chambers et al, 2001; Guyon et al, 2003b).

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 28 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Several approaches have been used to create zebrafish models of human disease. One is mutagenesis
via immersion of male zebrafish in ethylnitrosurea followed by crossing with wild-type females, which
created a large number of mutant strains (Haffter et al, 1996). A phenotype screen of these mutants
yielded 166 strains with defects in embryonic motility, of which 63 (representing 18 distinct genes) also
had defects in skeletal muscle architecture (Granato et al, 1996). The mutation in one of these strains,
sapje (sap), was localized in dmd, the zebrafish homologue of human DMD (Bassett et al, 2003;
Bolanos-Jimenez et al, 2001). These fish exhibit progressive skeletal muscle degeneration due to
detachment of muscle fibers (Bassett et al, 2003).
A second approach to creating zebrafish models of disease is morpholino inhibition of target genes. This
has been performed on dystrophin (Guyon et al, 2003b), dystroglycan (Parsons et al, 2002), and
-sarcoglycan (Guyon et al, 2005). The resulting fish displayed decreased motility, and their spines were
bent or curved rather than straight (Fig. 216-4).

Dystrophin-null sapje mutant zebrafish (bottom) have patchy muscle birefringence at 7 days
postfertilization compared with wild-type zebrafish (top). Birefringence measures the amount of polarized
light that is rotated by highly ordered skeletal muscle. Patchy birefringence indicates muscle tearing in the
sapje mutant (Bassett et al, 2003). (Courtesy of J. Guyon and T. Pusack.)
These observations confirm that zebrafish can exhibit dystrophic phenotypes and be used as animal
models of muscular dystrophy, especially when large populations need to be studied.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 29 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Diagnosis
Serum Muscle Enzyme Measurements
Until the mid-twentieth century, muscular dystrophy was diagnosed entirely by clinical and pathologic
criteria. Aldolase, a glycolytic enzyme, was the first serum marker of muscular dystrophy identified (Sibley,
Lehninger, 1949). However, elevations in aldolase are also found in certain liver diseases and neoplasms
(Okinaka et al, 1961). The most specific serum marker for muscular dystrophy is creatine phosphokinase
(CPK or CK) (Ebashi et al, 1959; Okinaka et al, 1961). Several enzymes that are predominantly
expressed in liver are also found in muscle, but they are relatively nonspecific markers: alanine
aminotransferase (ALT, previously SGPT), aspartate aminotransferase (AST, previously SGOT), and
(lactate dehydrogenase (LDH) (Soltan, Blanchaer, 1959).
Measurements of serum markers, especially creatine phosphokinase and aldolase, are extremely useful
in screening patients with muscle weakness. An extremely high CK level (in the thousands of units per
liter) is highly suggestive of a muscular dystrophy (Dreyfus et al, 1960; Ebashi et al, 1959; Okinaka et al,
1961; Okinaka et al, 1959). Normal results or mild elevations (in the hundreds) may be consistent with a
handful of muscular dystrophies such as FSHD but often suggest another etiology. If the patient has true
proximal muscle weakness and depressed or absent reflexes, the possibility of spinal muscular atrophy
should be considered in the setting of normal or wildly elevated CK levels (type III, Kugelberg Welander, in
patients who are ambulatory). If the patient has no verifiable weakness, normal levels help to exclude the
possibility of muscular dystrophy.
In some cases, an asymptomatic or mildly symptomatic child with an undiagnosed case of muscular
dystrophy has liver enzymes tested for an unrelated medical issue, and may go through an evaluation for
hepatic disease when mild elevations in ALT, AST, and LDH are identified. It is important for general
pediatricians and pediatric gastroenterologists to remember that these enzymes may be mildly elevated in
cases of muscular dystrophy, and checking a creatine phosphokinase level in this situation may save the
child a prolonged evaluation.
Nerve Conduction Studies and Electromyography
Electromyography (EMG) is a useful diagnostic tool in many types of neuromuscular disorders, but it is
rarely used anymore in the evaluation of possible muscular dystrophies. The combination of muscle
enzyme measurements, genetic testing, and muscle pathology has formed a powerful diagnostic triad that
is extremely accurate. Before genetic testing and immunohistochemistry were developed, myopathic
findings on EMG were used to support the diagnosis of muscular dystrophy.
Genetic Testing
Genetic testing for many common and rare muscular dystrophies is becoming increasingly available from
a number of sources. The website http://www.genetests.org is a useful resource to determine whether
genetic testing is available for a particular disorder.
Relatively simple multiplex PCR amplification of "hotspot" exons, including 2 to 20 and 44 to 53, can
detect single-exon and multiexon deletions that are responsible for 65% of all cases of DMD and BMD
(Figs. 216-5 and 216-6) (Beggs et al, 1990; Chamberlain et al, 1988a; Chamberlain et al, 1991; Oudet et
al, 1992). Among the other mutations, 5% are duplications and 30% point mutations or small-scale
deletions or duplications (Galvagni et al, 1994; Hu et al, 1988; Hu et al, 1990; Roberts et al, 1994). The
DNA required for this testing can be extracted from blood lymphocytes. This has made the diagnostic

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 30 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

evaluation of muscular dystrophy much less invasive for those boys who present with a classic DMD or
BMD phenotype. If such a child has an elevated CK level and a positive genetic test, the diagnosis has
been established and muscle biopsy is not required unless the family and physician are interested in
quantifying the level of dystrophin expression in the muscle tissue in an attempt to refine the prognosis.
Typically, out-of-frame or nonsense mutations are associated with DMD, while in-frame mutations are
associated with BMD, but there are exceptions to this rule (Hu et al, 1992; Malhotra et al, 1988).

Schematic diagram showing the spectrum of deletions (thin bars) and duplications (thick bars) relative to
the position of exons in the mRNA and to the dystrophin molecule. Each bar represents the extent of the
deletion or duplication in a single patient. The hot spot for deletion around 45-52 is evident, as is the
collection of larger deletions found in the 5 half of the gene.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 31 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Agarose gel electrophoresis of multiplex polymerase chain reaction products from blood cell DNA of four
patients. Two multiplex reactions are shown. The first set of four lanes contains amplified products of
exons 48, 17, 8, and 44; the second set of four lanes contains amplified products of exons 45, 19, 25, 12,
and 4. In patient 1, exons 48-52 are deleted, and in patient 3, exons 45-52 are deleted. In patient 2 and in
two normal controls (...
It is a difficult situation when the genetic test is negative in a boy with weakness and an elevated CK level.
Some clinicians will request or perform a muscle biopsy at this point, whereas others will pursue
sequencing of the entire DMD gene in an attempt to spare the child a muscle biopsy. The advantage of
examining muscle pathology is that it casts a wide net and can detect evidence of a wide variety of muscle
and nerve disorders. The disadvantage is its invasive nature. The advantage of DMD sequencing is that it
is noninvasive, as it requires only DNA obtained from blood lymphocytes. However, it is currently
expensive, and it will not detect evidence for other disorders such as the LGMDs. This decision should be
made based on the level of clinical suspicion for DMD or BMD, and the availability of muscle biopsy and
pathology facilities.
Carrier testing of parents of affected patients is important. In cases of DMD or BMD, only the mother
typically requires analysis. This can be performed by laser densitometry or fluorescence in situ
hybridization (FISH) techniques (Allingham-Hawkins et al, 1998; Calvano et al, 1997). A complicating
factor in DMD/BMD genetics is the possibility that the mother may be a carrier with germ-line mosaicism,
and thus testing of DNA obtained from blood leukocytes or other somatic cells will not reveal the mutation.
Thus, when the mother tests negative as a carrier, the brother of a DMD/BMD patient who inherits the
same X chromosome as his brother has a 10 to 30% chance of also developing DMD/BMD (Bakker et al,
1989c; Bullock et al, 1996; Grimm et al, 1994; van Essen et al, 1992). Prenatal testing should be
considered even when the mother does not initially appear to be a carrier (Bakker et al, 1989a; Bakker et
al, 1989b). Fetal cells may be obtained for analysis using amniocentesis or by extracting fetal nucleated
erythrocytes from maternal blood samples (Sekizawa et al, 1996). Preimplantation diagnosis can help

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 32 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

screen for unaffected embryos at the very beginning of gestation (Grifo et al, 1994; Kristjansson et al,
1994; Liu et al, 1995).
Carrier testing is also possible in other muscular dystrophies such as merosin-deficient congenital
muscular dystrophy, using methods such as linkage analysis (if only the linkage region is known),
mutation analysis (if the gene is known and the mutation has been identified in the proband), and
expression analysis (if protein expression is detectable and reliable) (Guicheney et al, 1998; Naom et al,
1997a; Naom et al, 1997b; Voit et al, 1994).
Occasionally, an asymptomatic female may present with an elevated CK level. The most likely explanation
is that she is a carrier for DMD or BMD, and it may be worth performing genetic testing before pursuing a
muscle biopsy.
More recently, PCR primers designed to operate under the same conditions for all 79 exons of DMD have
been developed. These primers generate amplicons approximately 1000 bp in length. Internal primers are
then used for sequencing (Flanigan et al, 2003). An alternative approach uses automated primer design
tools to extend this to other genes involved in muscular dystrophy (Bennett et al, 2004). Clinical
implementation of these techniques will further refine the diagnostic accuracy of the muscular dystrophies.
This is a sorely needed advance, as the rate of misdiagnosis or absence of a molecular diagnosis for the
muscular dystrophies remains disturbingly high nearly two decades after the gene for DMD was cloned
(Griggs, Bushby, 2005; Schwartz et al, 2005).
Muscle Pathology
The pathologic diagnosis of the muscular dystrophies was not very specific before the DMD gene was
cloned in 1986, since the biochemical defect was not previously known. Standard histochemical stains
such as hematoxylin and eosin demonstrated infiltration of inflammatory cells, necrosis, regeneration of
fibers, and, in the later stages, replacement of muscle fibers with connective and adipose tissues (Figs.
216-7 and 216-8). These changes are suggestive of muscular dystrophy but do not provide any indication
of the subtype. Additionally, some cases of inflammatory myopathy may resemble some cases of
muscular dystrophy in basic histology, and conversely, also detracting from the utility of basic
histochemical stains in isolation.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 33 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Cryostat section of an early-stage DMD muscle biopsy. Several necrotic fibers undergo phagocytosis.
There is an abnormal variability of the muscle fiber size but relatively little endomysial connective tissue
excess (modified trichrome stain; 350).

Cryostat section of an advanced-stage DMD muscle biopsy. A group of necrotic and regenerating muscle
fibers is apparent. In addition, there is a marked variability of fiber size and conspicuous excess of
endomysial connective tissue (modified trichrome stain; 350).

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 34 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

After the protein product, dystrophin, was identified, antibodies against various portions of the protein
were developed and used with fluorescence or radioactive isotopes to quantify (immunoblots, also known
as Western blots) and localize it (immunohistochemistry) within the muscle fiber. Clinically, immunoblots
and immunohistochemical stains could be used to determine the presence or absence of dystrophin within
the muscle and thus provide a means of diagnosis at a time when DNA sequencing was still a laborious
and difficult process (Fig. 216-9) (Arahata et al, 1988; Bonilla et al, 1988; Zubrzycka-Gaarn et al, 1988). In
most cases of DMD, dystrophin is virtually undetectable (less than 3% of normal levels) on immunoblot
analysis, while in BMD dystrophin is present at 40 to 100% of normal levels but typically has an abnormal
molecular weight (Fig. 216-10) (Arahata et al, 1989; Hoffman et al, 1988a). Manifesting female carriers will
have a partial deficiency of dystrophin (Fig. 216-11).

A, B, Transverse cryostat sections of muscle showing immunoreactive dystrophin using a monoclonal


antibody recognizing rod-domain epitopes. The bound antibodies are displayed by peroxidase reaction. A,
Normal muscle. B, Muscle from DMD patient. C, D, Serial sections from a DMD muscle biopsy. C,
Antidystrophin antibody as in A and B displayed by CI3 fluorochrome. D, Antiutrophin monoclonal
antibody for the N-terminus. In C, "revert...

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 35 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Immunoblot (left) showing normal-sized 427-kDa dystrophin from a normal muscle biopsy (N) and a
lower-molecular-weight dystrophin in reduced amount from a BMD patient (B). Muscle biopsy from the
same BMD patient (right) shows weak and interrupted dystrophin immunoperoxidase staining (350).

Dystrophin immunofluorescence from a manifesting carrier showing a cluster of dystrophin-negative


muscle fibers that had normal spectrin and laminin immunostaining (350).
In recent years, antibodies to a number of other muscle proteins have been developed, including the
sarcoglycans, merosin, and emerin, enabling pathologists to stain muscle samples with a panel of
antibodies to narrow the diagnostic possibilities (Figs. 216-12, 216-13, and 216-14). To interpret a pattern
of immunohistochemical stains, it is important to be familiar with the interactions between proteins. A
deficient pattern of staining in one protein does not always imply that the corresponding gene is
responsible for the dystrophy.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 36 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Immunostaining with antibody against -sarcoglycan in muscle sections from (A) a 13-year-old boy who
presented with a Duchenne-like muscular dystrophy and (B) a normal control. The patient was shown to
have a missense mutation in the -sarcoglycan gene, resulting in failure to integrate into the complex.

Immunostaining with antimerosin antibody of muscle from (A) merosin-deficient patient with congenital
muscular dystrophy and (B) normal control. In the patient muscle, a few fibers retain some
immunoreactive merosin (350).

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 37 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Immunostaining with antiemerin antibody of muscle from (A) normal control and (B) a patient with
Emery-Dreifuss muscular dystrophy. In the control tissue, emerin is localized to the membrane of
myonuclei (350).
In DMD, there is a secondary deficiency of the dystroglycans and sarcoglycans and normal merosin
staining (Ohlendieck et al, 1993). In another example, a mutation in a sarcoglycan gene will typically
cause not only an absence of staining for the corresponding protein, but also partial deficiencies in one or
more of the other sarcoglycans. This occurs most consistently in - and -sarcoglycanopathy (Vainzof et
al, 1996). The secondary effects of - and -sarcoglycanopathies on the other sarcoglycans and
sarcospan are less consistent, and sometimes there may be no secondary biochemical effects at all
(Crosbie et al, 1997; Crosbie et al, 2000). Primary -sarcoglycanopathy may in some cases be
accompanied by secondary deficiencies in - and -sarcoglycans (Barresi et al, 1997). Dystrophin staining
is usually normal in the sarcoglycanopathies (Ljunggren et al, 1995). In light of these secondary effects,
immunohistochemical analysis of the sarcoglycans in muscle specimens obtained for diagnostic purposes
should include all four for a proper interpretation. A diagnosis of an inherited disorder such as muscular
dystrophy is never completely established unless a mutation is found in a causative gene. The interactions
among various muscle proteins raise the possibility that in the future, synergistic heterozygosity will be
found to cause rare cases of muscular dystrophy in which traditional mutations are not identified. This
phenomenon arises from partial defects at multiple points in a pathway, and has been postulated to occur
in some metabolic muscle disorders (Vockley et al, 2000).

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 38 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Therapies
Because it is by far the most common muscular dystrophy, DMD has received the most attention for
therapeutic studies, whether performed in vitro, in animal models, or in human clinical trials. The therapy
for DMD is the most highly developed. Therapies for other muscular dystrophies are largely derived from
experience with DMD. Despite the significant advances in supportive therapies, including surgeries and
mechanical ventilation, DMD remains a fatal disease, and intense and diverse efforts to find a cure remain
quite active.
Patients and their families should be warned of two potentially serious acute muscular complications of
certain myopathies that require emergency medical evaluation: malignant hyperthermia and
myoglobinuria/rhabdomyolysis. Primary malignant hyperthermia is a separate disease category,
associated with mutations in various genes, most prominently RYR1, the ryanodine receptor (MacLennan
et al, 1990; McCarthy et al, 1990). Secondary malignant hyperthermia may occur in association with
certain myopathies, most commonly central core disease (also associated with mutations in RYR1), but
also with muscular dystrophies and other muscle disorders. This condition is typically triggered by specific
anesthetic agents, most notably succinylcholine and halothane; inhaled agents other than halothane
should also be avoided. The syndrome is potentially fatal and is characterized by severe muscle rigidity,
myoglobinuria/rhabdomyolysis, hyperthermia, and hyperkalemia. Dantrolene is the key therapy;
supportive treatment of the myoglobinuria/rhabdomyolysis, hyperthermia, and hyperkalemia are also
critical for survival. Myoglobinuria/rhabdomyolysis in isolation may also occur in muscle disorders and may
be triggered by environmental stressors such as high or low temperatures, excessive exercise, infection,
toxins, alcohol, and trauma. Any electrolyte imbalances associated with this condition should be treated
immediately, and intravenous fluids will often be required to prevent kidney damage.
Without any specific therapy, the life expectancy of a child with DMD does not typically extend beyond the
second decade of life. However, supportive therapies have significantly improved the quality of life and life
expectancy, to the point where most patients survive well into their third and sometimes even fourth
decade (Eagle et al, 2002). New therapies have been developed to reduce the impact of both the
pulmonary and cardiac complications, by far the most common causes of death in affected children. Aside
from a pediatric neurologist, other physicians frequently involved in the care of a child with DMD include
an orthopedic surgeon, pulmonologist, and cardiologist. Physical therapists also contribute a great deal to
the care of these children.
Steroid Therapy
Prednisones potential as a therapy for DMD was first raised in a pilot study of 14 patients that was
published in 1974 (Drachman et al, 1974). This approach was somewhat controversial, and the very
obvious side effects of prednisone made it difficult to conduct a blind, placebo-controlled trial, although a
placebo-controlled trial without blinding was feasible. Because a large number of DMD patients follow a
predictable course, some investigators in the 1970s and 1980s began to establish a baseline natural
history against which therapeutic trials could be measured (Mendell et al, 1987). A clinical trial of
prednisone in 33 boys affected by DMD was conducted using these natural history controls,
demonstrating improvement in strength over a 6-month period (Brooke et al, 1987).
The first long-term trial of prednisone, at a dose of 2 mg/kg per day in 16 affected boys, demonstrated that
the mean age of loss of ambulation could be delayed from 10 years (range, 7-13 years) to 12 years, a
significant gain in quality of life (DeSilva et al, 1987). A larger trial divided 103 boys with DMD into three
treatment groups: 1.5 mg/kg per day prednisone, 0.75 mg/kg per day prednisone, and placebo. The boys
in both prednisone groups experienced significant improvements in multiple measures of strength and

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 39 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

function over the 6-month trial period, suggesting that the lower dose may be sufficient (Mendell et al,
1989). A trial involving 93 affected boys demonstrated that a dose of 0.75 mg/kg per day was efficacious
(Fenichel et al, 1991a). Since then, several randomized, controlled clinical trials lasting 6 to 18 months
have been conducted, confirming an optimal prednisone dose of 0.75 mg/kg per day (maximum, 60-80
mg/d) to improve motor strength and pulmonary function (Griggs et al, 1991; Griggs et al, 1993).
Deflazacort has been explored as an alternative steroid to prednisone (Angelini et al, 1994; Mesa et al,
1991; Reitter, 1995). At a dose of 0.9 mg/kg per day, it provides similar benefits, with one small study
demonstrating milder side effects than prednisone (Bonifati et al, 2000).
Steroid therapy has become increasingly accepted as an efficacious therapy for DMD and potentially for
other muscular dystrophies. However, a trial in FSHD did not demonstrate a beneficial effect (Tawil et al,
1997), and thus steroids are not routinely used in other forms of muscular dystrophy.
Unfortunately, side effects of steroid therapy are numerous and potentially severe. Reported side effects
in DMD patients treated with prednisone include weight gain, stress fracture due to osteopenia,
hyperactivity and other behavioral changes, gastritis, hypertension, cataracts, and
hyperglycemia/glycosuria (DeSilva et al, 1987; Fenichel et al, 1991a). The effect of the weight gain is
unclear, as some of the additional weight may be a result of increased muscle mass, but excessive weight
gain due to adipose tissue may be counterproductive by increasing the load borne by the musculoskeletal
system without improving strength. Osteopenia increases the risk of fractures in children who already fall
more frequently than their peers.
To reduce the incidence and severity of steroid side effects, variations on the standard regimen have been
studied, including ones involving lower doses (0.3 mg/kg per day) and ones in which the steroids are not
administered on a daily basis. Common schedules include every-other-day dosing, which initially
appeared to be less efficacious and to carry the same side effect risks as daily dosing (Fenichel et al,
1991b). These schedules provide slightly less efficacious therapy but do alleviate the side effects. More
thorough evaluations of various alternate regimens are needed (Moxley et al, 2005).
The mechanism of action of steroids in the setting of DMD is not entirely clear, although the
anti-inflammatory effect appears to contribute. Prednisone was demonstrated to increase dystrophin
expression in myoblast cultures (Sklar, Brown, 1991), but this could not be confirmed in pre- and
posttreatment muscle biopsies in humans (Burrow et al, 1991). A decrease in inflammatory activity was,
however, noted in human muscle biopsies, suggesting that the anti-inflammatory properties of prednisone
are key to its efficacy (Kissel et al, 1991). The immunosuppressive effect does not appear to be the
primary mechanism of action (Kissel et al, 1993). In cell culture, steroids promote various aspects of
myogenesis (Passaquin et al, 1993), including the fusion of myoblasts (Metzinger et al, 1993) and
prevention of myotube death (Sklar, Brown, 1991). Steroids have also been shown to alleviate myofiber
damage in rats (Jacobs et al, 1996). Metabolic studies in humans suggest that steroids improve muscle
mass by inhibiting proteolysis (Rifai et al, 1995).
-Agonist Therapy
Administration of albuterol has been shown to increase muscle mass and some measures of strength in
patients with facioscapulohumeral muscular dystrophy, but the improvement overall was not dramatic
(Kissel et al, 1998; Kissel et al, 2001).

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 40 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Supportive Management
There are a number of orthopedic and orthotic interventions that apply to DMD. The one that perhaps has
had the greatest impact is scoliosis surgery. Scoliosis is a complication that typically accelerates after the
child begins sitting in a wheelchair for long periods of time. Not only does scoliosis cause discomfort and
social embarrassment due to postural changes, but the imbalances in lung volumes increase the
likelihood of developing atelectasis, resulting in reduced lung capacities and more frequent episodes of
pneumonia. The scoliosis surgery stabilizes the spine and enables the child to breathe more normally,
reducing the impact of potentially fatal pulmonary complications.
Due to the high risk of pneumonias and respiratory compromise in advanced cases, affected children will
need to have their lung function monitored with pulmonary function testing (PFTs), and a pulmonologist
should be consulted. Respiratory infections should be treated more aggressively than in unaffected
children, and the threshold for hospital admission should be lower for such complications.
Various options for mechanical ventilation have been explored over the past several decades, and its use
has become routine in the later stages of the disease. A number of studies have suggested that
mechanical ventilation prolongs life expectancy, including two recently published retrospective
observations covering several decades (Eagle et al, 2002; Jeppesen et al, 2003). A less invasive
alternative method, nocturnal nasal ventilation, may be helpful in selected patients (Guilleminault et al,
1998; Simonds et al, 1998).
The cardiac complications of the muscular dystrophies include cardiomyopathy and arrhythmias.
Previously, no specific therapy was available for these complications, and thus routine monitoring of
asymptomatic individuals was not common. However, interventions have become more sophisticated,
including the use of ACE inhibitors and beta blockers for cardiomyopathy. Thus, patients newly diagnosed
with a muscular dystrophy are now commonly referred for a baseline cardiac evaluation, including
electrocardiography and echocardiography, and followed at increasingly frequent intervals as the disease
progresses (Bushby et al, 2003). However, there is some disagreement about the utility of monitoring
asymptomatic individuals (English, Gibbs, 2006). Cardiac transplantation is generally not feasible in DMD
but may be a consideration in selected cases of BMD (Bushby et al, 2003; English, Gibbs, 2006).
When heel cord contractures develop, the least invasive therapies include stretching exercises under the
guidance of a physical therapist. In more severe cases, the exercises may be supplemented by orthotic
devices such as molded ankle-foot orthoses (MAFOs) or night casts. In the most severe cases, especially
when the contractures limit the ability of the patient to walk, heel cord lengthening surgery may be
beneficial. However, this surgery is not a permanent solution, and over time the contractures may worsen
again.
Mild to moderate mental retardation is a frequent, though not universal, complication of DMD. Autistic
spectrum phenotypes also have been observed in cases of DMD and occur with a greater frequency than
in the background population (Wu et al, 2005). Children with DMD should in general not be placed in
special education settings based on their physical disabilities alone, but if cognitive or behavioral
disabilities are significant, such programs may be appropriate for certain children.
The overall goal of therapy is to maintain mobility as long as possible. Within reasonable limits, exercise
and physical activity are beneficial, and affected children should be encouraged to remain active within
their families and communities.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 41 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Experimental Therapies
As soon as the DMD gene was cloned, scientists and physicians began to envision genetically targeted
therapies for the disease. Over the past two decades, a number of approaches have been studied that
propose different solutions to four key issues: what variant of DMD to insert, what vector to use, whether
to use cell-based or non-cell-based approaches (and if so, what type of cell), and mode of delivery.
Direct gene therapy was explored early. In one set of experiments, full-length murine dystrophin cDNA
was attached to an M-creatine kinase promoter enhancer region and then directly injected into mdx
mouse zygotes (Cox et al, 1993).
Gene Therapy
The use of viral vectors has been explored extensively. Their use is complicated by the enormous size of
the DMD gene. Full-length DMD will not fit in most viral vectors. Researchers developed truncated
dystrophin constructs that could fit into a viral vector (Acsadi et al, 1991) and correct some of the
pathologic features of dystrophin deficiency in mdx mice (Wells et al, 1992). Adenovirus has been the
easiest vector to work with due to its efficient delivery system (Graham et al, 1977). Expression of a
Becker minigene has been demonstrated in mdx mice (Acsadi et al, 1996; Clemens et al, 1995; Ragot et
al, 1994), -sarcoglycan in the BIO 14.6 hamster (Holt et al, 1998), and -sarcoglycan in a knockout
mouse (Duclos et al, 1998). However, the duration of expression was brief in all of these experiments.
Adeno-associated virus (AAV) has shown promise as an alternative vector. It is much smaller than
adenovirus and is less prone to triggering an immune response in the host (Xiao et al, 1996). The main
drawback is its limited capacity of 5 kb; the human dystrophin cDNA is 14 kb long. For this reason, one of
the first muscular dystrophy genes replaced using this vector was SGCD (-sarcoglycan), a smaller gene
that was delivered by intramuscular injection to the BIO14.6 dystrophic hamster with restoration of
histologic structures (Li et al, 1999) and functional recovery of muscle (Xiao et al, 2000). Administration of
AAV carrying SGCD into the coronary arteries of the BIO14.6 hamster resulted in histologic improvement
of the cardiac muscle (Li et al, 2003). More recently, intraperitoneal delivery of AAV-8 containing SGCD
into TO-2 hamsters, which are derivatives of the BIO14.6 strain, led to histologic and functional recovery
of both skeletal and cardiac muscle (Zhu et al, 2005).
In an attempt to address dystrophin deficiency using the AAV vector, several minidystrophin genes
smaller than 4.2 kb were constructed and delivered to mdx mice (Wang et al, 2000). Two of the
constructs, 3849 and 3990, were able to restore the DAPC architecture on a histologic level. Other
truncated dystrophin constructs, more recently termed microdystrophins, also have been studied in the
context of the AAV vector. Delivery of a R4 microdystrophin to the heart of a newborn mdx mouse
produced restoration of the DAPC in the heart (Yue et al, 2003). Another construct, CS1, was injected
into the tibialis anterior muscles of mdx mice, which were found to have improved force generation
(Yoshimura et al, 2004). Intramuscular injections of AAV-5 containing a R4/C microdystrophin were
more effective in young rather than adult mdx mice; the young mice displayed some histologic and
functional improvements in the injected muscles (Liu et al, 2005). Intramuscular injections of AAV-6 vector
containing the H2-R19 minidystrophin gene led to moderate functional improvement (Lai et al, 2005).
Systemic injection of AAV pseudotype 6 vector containing R4-23/CT microdystrophin into the tail vein
of mdx mice resulted in widespread expression of the microdystrophin in skeletal muscles, with some
functional improvement (Gregorevic et al, 2004).

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 42 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Cell-Based Therapies
Restoration of DMD expression via cell-based therapies has been explored extensively since the late
1980s. One approach adapts the muscles ability to repair itself using satellite cells, which are muscle
precursor cells found between the sarcolemmal and basement membranes of muscles (Seale, Rudnicki,
2000). In one experiment, dystrophin expression was induced by direct injection of muscle precursor cells
into affected muscles of the mdx mouse (Partridge et al, 1989). When a similar approach was studied in
human patients with DMD, dystrophin expression was detected (Gussoni et al, 1992), but improvements
in strength found in a very small sample size were only transient (Huard et al, 1992a) and the overall
results were judged to be poor (Karpati et al, 1993a; Mendell et al, 1995; Miller et al, 1997; Tremblay et al,
1993). Further examination of muscle tissue from study subjects suggested that incomplete fusion of
myoblasts with host muscle fibers contributed to the unsatisfactory outcome (Gussoni et al, 1997; Gussoni
et al, 1996).
It then became apparent that the mononuclear cell population in muscle was heterogeneous and that
different subpopulations might have greater myogenic potential than others (Beauchamp et al, 1999; Qu
et al, 1998). Further research was directed at devising techniques for segregating these subpopulations
and identifying which ones had the greatest myogenic potential.
Bone marrow-derived mesenchymal cells are recruited to assist in skeletal muscle repair after injury,
suggesting that a subpopulation of these cells has myogenic potential (Cossu, 1997; Ferrari et al, 1998).
Bone marrow transplantation in mdx mice resulted in the presence of bone marrow-derived cells within
skeletal muscle (Bittner et al, 1999).
Muscle is also a potential source of precursor cells. A subpopulation of hematopoietic stem cells shared
the ability to expel Hoechst 33342 dye at a high rate, enabling them to be isolated from other
hematopoietic stem cells (Goodell et al, 1996). This technique was adapted to isolate a side population
(SP) of muscle-derived cells (Fig. 216-15) (Gussoni et al, 1999) that demonstrate multilinear engraftment
in mice (Jackson et al, 1999) and promote bone healing (Lee et al, 2000). When injected intravenously
into mdx (Gussoni et al, 1999) and mdx 5cv (Bachrach et al, 2004) mice, these SP cells partially restored
dystrophin expression in skeletal muscle.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 43 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Immunostaining of cultured side population (SP) and main population (MP) cells derived from mouse
muscle. Cultured SP cells stain positive for the chemokine receptor CXCR4 (arrows) using two
monoclonal antibodies: (A) 12G5 conjugated to phycoerythrin (PE) and (C) 2B11 conjugated to
fluorescein isothiocyanate (FITC). In contrast, cultured MP cells do not express CXCR4 (B and D).
(Courtesy of E. Bachrach and E. Luth.)
There are other potential sources of muscle precursor cells, including umbilical cord blood, peripheral
blood, and brain (Bjornson et al, 1999; Clarke et al, 2000; Lu et al, 1996).
The optimal mode of delivery has been controversial, and this issue has been complicated by the choices
made to address the other issues above. Intramuscular and intravenous injections have been most
commonly used. The disadvantage of intramuscular injections is the limited diffusion capacity of DNA,
viruses, or muscle precursor cells within the muscle itself outside the vasculature. Intravenous injections
allow for more diffuse seeding of the muscle, but the disadvantage, especially with cell-based therapies, is
that the cells will be filtered by the lungs, reducing the number that reach the skeletal muscles themselves.
An alternative mode of delivery is via intra-arterial injections directly into the iliac artery (mice) or femoral
artery (humans). The former has been attempted in -sarcoglycan-deficient mice (Sampaolesi et al, 2003)
and mdx 5cv mice (Bachrach et al, 2006) with promising results.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 44 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Exon Skipping
Another genetic approach to treating genetic disorders involves exon skipping, or inducing the
transcriptional mechanism of the nucleus to skip over a particular exon. This may be achieved by
administering 2-O-methyl antisense oligoribonucleotides that bind to the splice site of the targeted exon
(Dominski, Kole, 1993). When a frameshift or nonsense mutation is present in an exon, this intervention
can enable the cell to produce a mildly truncated but functional protein rather than a severely truncated
protein that becomes degraded or is nonfunctional. Exon 23 of the murine version of DMD in the mdx
mouse was targeted in this manner via intramuscular injections, producing intact production and
localization of dystrophin in muscle fibers (Mann et al, 2001). Further study of mice treated in this fashion
demonstrated functional improvement in muscle strength (Lu et al, 2003). Systemic injection of these
oligonucleotides into the tail vein of mdx mice demonstrated restored dystrophin expression in skeletal
muscles, but at widely varying levels, and there was no uptake at all in the heart (Lu et al, 2005). Systemic
intravenous delivery of an alternative morpholino oligonucleotide in mdx mice also produced a wide range
of expression levels among skeletal muscles, but pathologic and functional improvement was observed
(Alter et al, 2006).
Myostatin Inhibition
Myostatin, also known as growth/differentiation factor 8, is an inhibitor of muscle growth that has been
studied for some time (McPherron et al, 1997). Mice (McPherron et al, 1997; Zhu et al, 2000) and cattle
(Grobet et al, 1997; Grobet et al, 1998; Kambadur et al, 1997; McPherron, Lee, 1997) with mutations in
the myostatin gene were noted to have dramatically increased muscle mass. Inhibition of myostatin in
wild-type (Whittemore et al, 2003) and mdx (Bogdanovich et al, 2002) mice via intraperitoneal injection of
monoclonal antibodies resulted in increased muscle mass and strength. To date, only one human has
been found to have a naturally occurring mutation in myostatin (Schuelke et al, 2004). A boy, last
examined at 4.5 years, had a homozygous splice-site mutation in the myostatin gene, with a phenotype of
increased muscle mass and motor strength (Schuelke et al, 2004). It was especially encouraging that at
that age, he had not developed any theoretical complications of myostatin deficiency, especially
hypertrophic cardiomyopathy; however, further observation of this child over time will provide more
important information about the effects of myostatin deficiency in humans.
Conclusion
The scientific and clinical advances in the muscular dystrophies over the past several decades have been
remarkable, with important discoveries in the genetic, biochemical, and therapeutic aspects of the field. A
precise molecular diagnosis of a large number of muscular dystrophies is now possible, and therapeutic
refinements have dramatically increased the life expectancy and quality of life in affected individuals.
Many of these patients and their families have great hopes that new therapies will eventually halt or even
reverse the progression of these diseases. Through a variety of approaches, including the ones outlined
above, scientists and physicians are making great efforts to fulfill those hopes.
Acknowledgments
The current authors are indebted to Ronald G. Worton, Maria Judit Molnar, Bernard Brais, and George
Karpati, authors of the previous version of this chapter. We have used a number of their figures, as well as
selected text and references. The authors are supported by NIH P01 NS40828-01A1 (L.M.K.), NIH K08
NS048180 (P.B.K.), the Bernard F. and Alva B. Gimbel Foundation (L.M.K.), and the Joshua Frase
Foundation (L.M.K.). L.M.K. is an Investigator at the Howard Hughes Medical Institute.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 45 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

REFERENCES
1. Diagnosis of Duchenne and Becker muscular dystrophies by polymerase chain reaction. A
multicenter study. JAMA. 267:2609, 1992. PMID 1573747
2. Achanzar WE, Ward S: A nematode gene required for sperm vesicle fusion. J Cell Sci. 110:1073,
1997. PMID 9175703
3. Acsadi G, Dickson G, Love DR, Jani A, Walsh FS, Gurusinghe A, Wolff JA, Davies KE: Human
dystrophin expression in mdx mice after intramuscular injection of DNA constructs. Nature. 352:815,
1991. PMID 1881437
4. Acsadi G, Lochmuller H, Jani A, Huard J, Massie B, Prescott S, Simoneau M, Petrof BJ, Karpati G:
Dystrophin expression in muscles of mdx mice after adenovirus-mediated in vivo gene transfer. Hum
Gene Ther. 7:129, 1996. PMID 8788164
5. Adams ME, Butler MH, Dwyer TM, Peters MF, Murnane AA, Froehner SC: Two forms of mouse
syntrophin, a 58 kd dystrophin-associated protein, differ in primary structure and tissue distribution.
Neuron. 11:531, 1993. PMID 7691103
6. Adams ME, Kramarcy N, Krall SP, Rossi SG, Rotundo RL, Sealock R, Froehner SC: Absence of
alpha-syntrophin leads to structurally aberrant neuromuscular synapses deficient in utrophin. J Cell
Biol. 150:1385, 2000. PMID 10995443
7. Ahn AH, Freener CA, Gussoni E, Yoshida M, Ozawa E, Kunkel LM: The three human syntrophin
genes are expressed in diverse tissues, have distinct chromosomal locations, and each bind to
dystrophin and its relatives. J Biol Chem. 271:2724, 1996. PMID 8576247
8. Ahn AH, Kunkel LM: Syntrophin binds to an alternatively spliced exon of dystrophin. J Cell Biol.
128:363, 1995. PMID 7844150
9. Ahn AH, Yoshida M, Anderson MS, Feener CA, Selig S, Hagiwara Y, Ozawa E, Kunkel LM: Cloning
of human basic A1, a distinct 59-kDa dystrophin-associated protein encoded on chromosome
8q23-24. Proc Natl Acad Sci USA. 91:4446, 1994. PMID 8183929
10. Aida N, Tamagawa K, Takada K, Yagishita A, Kobayashi N, Chikumaru K, Iwamoto H: Brain MR in
Fukuyama congenital muscular dystrophy. Am J Neuroradiol. 17:605, 1996. PMID 8730178
11. Aida N, Yagishita A, Takada K, Katsumata Y: Cerebellar MR in Fukuyama congenital muscular
dystrophy Polymicrogyria with cystic lesions. Am J Neuroradiol. 15:1755, 1994. PMID 7847224
12. Allingham-Hawkins DJ, McGlynn-Steele LK, Brown CA, Sutherland J, Ray PN: Impact of carrier
status determination for Duchenne/Becker muscular dystrophy by computer-assisted laser
densitometry. Am J Med Genet. 75:171, 1998. PMID 9450879
13. Alter J, Lou F, Rabinowitz A, Yin H, Rosenfeld J, Wilton SD, Partridge TA, Lu QL: Systemic delivery
of morpholino oligonucleotide restores dystrophin expression bodywide and improves dystrophic
pathology. Nat Med. 12:175, 2006. PMID 16444267
14. Anderson LV, Davison K, Moss JA, Richard I, Fardeau M, Tome FM, Hubner C, Lasa A, Colomer J,
Beckmann JS: Characterization of monoclonal antibodies to calpain 3 and protein expression in
muscle from patients with limb-girdle muscular dystrophy type 2A. Am J Pathol. 153:1169, 1998.
PMID 9777948
15. Angelini C, Fanin M, Menegazzo E, Freda MP, Duggan DJ, Hoffman EP: Homozygous
alpha-sarcoglycan mutation in two siblings One asymptomatic and one steroid-responsive mild
limb-girdle muscular dystrophy patient. Muscle Nerve. 21:769, 1998. PMID 9585331
16. Angelini C, Pegoraro E, Turella E, Intino MT, Pini A, Costa C: Deflazacort in Duchenne dystrophy
Study of long-term effect. Muscle Nerve. 17:386, 1994. PMID 8170484
17. Arahata K, Hoffman EP, Kunkel LM, Ishiura S, Tsukahara T, Ishihara T, Sunohara N, Nonaka I,
Ozawa E, Sugita H: Dystrophin diagnosis Comparison of dystrophin abnormalities by
immunofluorescence and immunoblot analyses. Proc Natl Acad Sci USA. 86:7154, 1989. PMID
2674948
18. Arahata K, Ishiura S, Ishiguro T, Tsukahara T, Suhara Y, Eguchi C, Ishihara T, Nonaka I, Ozawa E,

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 46 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

19.

20.

21.
22.

23.

24.

25.

26.

27.

28.

29.
30.
31.

32.

33.

34.

35.

Sugita H: Immunostaining of skeletal and cardiac muscle surface membrane with antibody against
Duchenne muscular dystrophy peptide. Nature. 333:861, 1988. PMID 3290683
Asmus F, Zimprich A, Tezenas Du Montcel S, Kabus C, Deuschl G, Kupsch A, Ziemann U, Castro M,
Kuhn AA, Strom TM, Vidailhet M, Bhatia KP, Durr A, Wood NW, Brice A, Gasser T: Myoclonus-dystonia
syndrome Epsilon-sarcoglycan mutations and phenotype. Ann Neurol. 52:489, 2002. PMID 12325078
Bachrach E, Li S, Perez AL, Schienda J, Liadaki K, Volinski J, Flint A, Chamberlain J, Kunkel LM:
Systemic delivery of human microdystrophin to regenerating mouse dystrophic muscle by muscle
progenitor cells. Proc Natl Acad Sci USA. 101:3581, 2004. PMID 14993597
Bachrach E: et al. : Muscle Nerve, 2006 (in press). PMID 14993597
Badorff C, Lee GH, Lamphear BJ, Martone ME, Campbell KP, Rhoads RE, Knowlton KU: Enteroviral
protease 2A cleaves dystrophin Evidence of cytoskeletal disruption in an acquired cardiomyopathy.
Nat Med. 5:320, 1999. PMID 10086389
Bakker E, Bonten EJ, den Dunnen JT, Veenema H, Grootscholten PM, van Ommen GJ, Pearson PL:
Carrier detection and prenatal diagnosis of Duchenne/Becker muscular dystrophy (D/BMD) by
DNA-analysis. Prog Clin Biol Res. 306:51, 1989a. PMID 2662213
Bakker E, Bonten EJ, Veenema H, den Dunnen JT, Grootscholten PM, van Ommen GJ, Pearson PL:
Prenatal diagnosis of Duchenne muscular dystrophy A three-year experience in a rapidly evolving field.
J Inherit Metab Dis 12(Suppl 1). : 174, 1989b. PMID 2509804
Bakker E, Hofker MH, Goor N, Mandel JL, Wrogemann K, Davies KE, Kunkel LM, Willard HF, Fenton
WA, Sandkuyl L: , et al Prenatal diagnosis and carrier detection of Duchenne muscular dystrophy with
closely linked RFLPs. JOURNALancetLTITLE. 1:655, 1985. PMID 2858615
Bakker E, Van der Wielen MJ, Voorhoeve E, Ippel PF, Padberg GW, Frants RR, Wijmenga C:
Diagnostic, predictive, and prenatal testing for facioscapulohumeral muscular dystrophy Diagnostic
approach for sporadic and familial cases. J Med Genet. 33:29, 1996. PMID 8825045
Bakker E, Veenema H, den Dunnen JT, van Broeckhoven C, Grootscholten PM, Bonten EJ, van
Ommen GJ, Pearson PL: Germinal mosaicism increases the recurrence risk for new Duchenne muscular
dystrophy mutations. J Med Genet. 26:553, 1989c. PMID 2810338
Balci B, Uyanik G, Dincer P, Gross C, Willer T, Talim B, Haliloglu G, Kale G, Hehr U, Winkler J,
Topaloglu H: An autosomal recessive limb girdle muscular dystrophy (LGMD2) with mild mental
retardation is allelic to Walker-Warburg syndrome (WWS) caused by a mutation in the POMT1 gene.
Neuromuscul Disord. 15:271, 2005. PMID 15792865
Bansal D, Miyake K, Vogel SS, Groh S, Chen CC, Williamson R, McNeil PL, Campbell KP: Defective
membrane repair in dysferlin-deficient muscular dystrophy. Nature. 423:168, 2003. PMID 12736685
Barohn RJ, Miller RG, Griggs RC: Autosomal recessive distal dystrophy. Neurology. 41:1365, 1991.
PMID 1891082
Barresi R, Confalonieri V, Lanfossi M, Di Blasi C, Torchiana E, Mantegazza R, Jarre L, Nardocci N,
Boffi P, Tezzon F, Pini A, Cornelio F, Mora M, Morandi L: Concomitant deficiency of beta- and
gamma-sarcoglycans in 20 alpha-sarcoglycan (adhalin)-deficient patients immunohistochemical analysis
and clinical aspects. Acta Neuropathol (Berl). 94:28, 1997. PMID 9224527
Bashir R, Britton S, Strachan T, Keers S, Vafiadaki E, Lako M, Richard I, Marchand S, Borg N,
Argov Z, Sadeh M, Mahjneh I, Marconi G, Passos-Bueno MR, Moreira Ede S, Zatz M, Beckmann JS,
Bushby K: A gene related to Caenorhabditis elegans spermatogenesis factor fer-1 is mutated in
limb-girdle muscular dystrophy type 2B. Nat Genet. 20:37, 1998. PMID 9731527
Bassett DI, Bryson-Richardson RJ, Daggett DF, Gautier P, Keenan DG, Currie PD: Dystrophin is
required for the formation of stable muscle attachments in the zebrafish embryo. Development.
130:5851, 2003. PMID 14573513
Beauchamp JR, Morgan JE, Pagel CN, Partridge TA: Dynamics of myoblast transplantation reveal a
discrete minority of precursors with stem cell-like properties as the myogenic source. J Cell Biol.
144:1113, 1999. PMID 10087257
Becker P: Dystrophia Musculorum Progressiva. Stuttgart, Thieme, 1953

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 47 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

36. Beggs AH, Koenig M, Boyce FM, Kunkel LM: Detection of 98% of DMD/BMD gene deletions by
polymerase chain reaction. Hum Genet. 86:45, 1990. PMID 2253937
37. Beltran-Valero de Bernabe D, Currier S, Steinbrecher A, Celli J, van Beusekom E, van der Zwaag B,
Kayserili H, Merlini L, Chitayat D, Dobyns WB, Cormand B, Lehesjoki AE, Cruces J, Voit T, Walsh CA,
van Bokhoven H, Brunner HG: Mutations in the O-mannosyltransferase gene POMT1 give rise to the
severe neuronal migration disorder Walker-Warburg syndrome. Am J Hum Genet. 71:1033, 2002.
PMID 12369018
38. Ben Jelloun-Dellagi S, Chaffey P, Hentati F, Ben Hamida C, Tome F, Colin H, Dellagi K, Kaplan JC,
Fardeau M, Ben Hamida M: Presence of normal dystrophin in Tunisian severe childhood autosomal
recessive muscular dystrophy. Neurology. 40:1903, 1990. PMID 2247244
39. Bennett RR, Schneider HE, Nutter RC, Lai PS, Barrett CE, Lip V, Wu BL, Darras BT, Kunkel LM:
Detection of muscular dystrophy genotypes via universal condition direct sequencing (UCDS). ASHG 54th
Annual Meeting Abstracts. Bethesda, American Society of Human Genetics, abstract 1485/F3.
Available online at http. : //www.ashg.org/genetics/ashg04s/
40. Bethlem J, Wijngaarden GK: Benign myopathy, with autosomal dominant inheritance. A report on
three pedigrees. Brain. 99:91, 1976. PMID 963533
41. Betz RC, Schoser BG, Kasper D, Ricker K, Ramirez A, Stein V, Torbergsen T, Lee YA, Nothen MM,
Wienker TF, Malin JP, Propping P, Reis A, Mortier W, Jentsch TJ, Vorgerd M, Kubisch C: Mutations in
CAV3 cause mechanical hyperirritability of skeletal muscle in rippling muscle disease. Nat Genet.
28:218, 2001. PMID 11431690
42. Bies RD, Friedman D, Roberts R, Perryman MB, Caskey CT: Expression and localization of
dystrophin in human cardiac Purkinje fibers. Circulation. 86:147, 1992. PMID 1617767
43. Bione S, Maestrini E, Rivella S, Mancini M, Regis S, Romeo G, Toniolo D: Identification of a novel
X-linked gene responsible for Emery-Dreifuss muscular dystrophy. Nat Genet. 8:323, 1994. PMID
7894480
44. Bione S, Small K, Aksmanovic VM, DUrso M, Ciccodicola A, Merlini L, Morandi L, Kress W, Yates
JR, Warren ST: , et al Identification of new mutations in the Emery-Dreifuss muscular dystrophy gene and
evidence for genetic heterogeneity of the disease. Hum Mol Genet. 4:1859, 1995. PMID 8595407
45. Bittner RE, Schofer C, Weipoltshammer K, Ivanova S, Streubel B, Hauser E, Freilinger M, Hoger H,
Elbe-Burger A, Wachtler F: Recruitment of bone-marrow-derived cells by skeletal and cardiac muscle in
adult dystrophic mdx mice. Anat Embryol (Berl). 199:391, 1999. PMID 10221450
46. Bjornson CR, Rietze RL, Reynolds BA, Magli MC, Vescovi AL: Turning brain into blood A
hematopoietic fate adopted by adult neural stem cells in vivo. Science. 283:534, 1999. PMID
9915700
47. Blumen SC, Nisipeanu P, Sadeh M, Asherov A, Blumen N, Wirguin Y, Khilkevich O, Carasso RL,
Korczyn AD: Epidemiology and inheritance of oculopharyngeal muscular dystrophy in Israel.
Neuromuscul Disord 7(Suppl 1). : S38, 1997. PMID 9392014
48. Blumen SC, Sadeh M, Korczyn AD, Rouche A, Nisipeanu P, Asherov A, Tome FM: Intranuclear
inclusions in oculopharyngeal muscular dystrophy among Bukhara Jews. Neurology. 46:1324, 1996.
PMID 8628475
49. Bogdanovich S, Krag TO, Barton ER, Morris LD, Whittemore LA, Ahima RS, Khurana TS: Functional
improvement of dystrophic muscle by myostatin blockade. Nature. 420:418, 2002. PMID 12459784
50. Boland BJ, Silbert PL, Groover RV, Wollan PC, Silverstein MD: Skeletal, cardiac, and smooth muscle
failure in Duchenne muscular dystrophy. Pediatr Neurol. 14:7, 1996. PMID 8652023
51. Bolanos-Jimenez F, Bordais A, Behra M, Strahle U, Sahel J, Rendon A: Dystrophin and Dp71, two
products of the DMD gene, show a different pattern of expression during embryonic development in
zebrafish. Mech Dev. 102:239, 2001. PMID 11287201
52. Bonifati MD, Ruzza G, Bonometto P, Berardinelli A, Gorni K, Orcesi S, Lanzi G, Angelini C: A
multicenter, double-blind, randomized trial of deflazacort versus prednisone in Duchenne muscular
dystrophy. Muscle Nerve. 23:1344, 2000. PMID 10951436

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 48 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

53. Bonilla E, Samitt CE, Miranda AF, Hays AP, Salviati G, DiMauro S, Kunkel LM, Hoffman EP, Rowland
LP: Duchenne muscular dystrophy Deficiency of dystrophin at the muscle cell surface. Cell. 54:447,
1988. PMID 3042151
54. Bonne G, Di Barletta MR, Varnous S, Becane HM, Hammouda EH, Merlini L, Muntoni F, Greenberg
CR, Gary F, Urtizberea JA, Duboc D, Fardeau M, Toniolo D, Schwartz K: Mutations in the gene encoding
lamin A/C cause autosomal dominant Emery-Dreifuss muscular dystrophy. Nat Genet. 21:285, 1999..
PMID 10080180
55. Bonnemann CG, Modi R, Noguchi S, Mizuno Y, Yoshida M, Gussoni E, McNally EM, Duggan DJ,
Angelini C, Hoffman EP: Beta-sarcoglycan (A3b) mutations cause autosomal recessive muscular
dystrophy with loss of the sarcoglycan complex. Nat Genet. 11:266, 1995. PMID 7581449
56. Bonnemann CG, Passos-Bueno MR, McNally EM, Vainzof M, de Sa Moreira E, Marie SK, Pavanello
RC, Noguchi S, Ozawa E, Zatz M, Kunkel LM: Genomic screening for beta-sarcoglycan gene mutations
Missense mutations may cause severe limb-girdle muscular dystrophy type 2E (LGMD 2E). Hum Mol
Genet. 5:1953, 1996. PMID 8968749
57. Bonnemann CG, Wong J, Ben Hamida C, Hamida MB, Hentati F, Kunkel LM: LGMD 2E in Tunisia is
caused by a homozygous missense mutation in beta-sarcoglycan exon 3. Neuromuscul Disord.
8:193, 1998. PMID 9631401
58. Bonnemann CG, Wong J, Jones KJ, Lidov HG, Feener CA, Shapiro F, Darras BT, Kunkel LM, North
KN: Primary gamma-sarcoglycanopathy (LGMD 2C) Broadening of the mutational spectrum guided by the
immunohistochemical profile. Neuromuscul Disord. 12:273, 2002. PMID 11801399
59. Bouchard JP, Brais B, Brunet D, Gould PV, Rouleau GA: Recent studies on oculopharyngeal
muscular dystrophy in Quebec. Neuromuscul Disord 7(Suppl 1). : S22, 1997. PMID 9392011
60. Bowe MA, Mendis DB, Fallon JR: The small leucine-rich repeat proteoglycan biglycan binds to
alpha-dystroglycan and is upregulated in dystrophic muscle. J Cell Biol. 148:801, 2000. PMID
10684260
61. Boyce FM, Beggs AH, Feener C, Kunkel LM: Dystrophin is transcribed in brain from a distant
upstream promoter. Proc Natl Acad Sci USA. 88:1276, 1991R. PMID 1996328
62. Boyd Y, Buckle V, Holt S, Munro E, Hunter D, Craig I: Muscular dystrophy in girls with X;autosome
translocations. J Med Genet. 23:484, 1986. PMID 3806636
63. Brais B, Bouchard JP, Gosselin F, Xie YG, Fardeau M, Tome FM, Rouleau GA: Using the full power
of linkage analysis in 11 French Canadian families to fine map the oculopharyngeal muscular dystrophy
gene. Neuromuscul Disord 7(Suppl 1). : S70, 1997. PMID 9392020
64. Brais B, Bouchard JP, Xie YG, Rochefort DL, Chretien N, Tome FM, Lafreniere RG, Rommens JM,
Uyama E, Nohira O, Blumen S, Korczyn AD, Heutink P, Mathieu J, Duranceau A, Codere F, Fardeau M,
Rouleau GA: Short GCG expansions in the PABP2 gene cause oculopharyngeal muscular dystrophy.
Nat Genet. 18:164, 1998. PMID 9462747
65. Brenman JE, Chao DS, Gee SH, McGee AW, Craven SE, SantillaNO DR, Wu Z, Huang F, Xia H,
Peters MF, Froehner SC, Bredt DS: Interaction of nitric oxide synthase with the postsynaptic density
protein PSD-95 and alpha1-syntrophin mediated by PDZ domains. Cell. 84:757, 1996. PMID 8625413
66. Brenman JE, Chao DS, Xia H, Aldape K, Bredt DS: Nitric oxide synthase complexed with dystrophin
and absent from skeletal muscle sarcolemma in Duchenne muscular dystrophy. Cell. 82:743, 1995.
PMID 7545544
67. Brockington M, Blake DJ, Prandini P, Brown SC, Torelli S, Benson MA, Ponting CP, Estournet B,
Romero NB, Mercuri E, Voit T, Sewry CA, Guicheney P, Muntoni F: Mutations in the fukutin-related
protein gene (FKRP) cause a form of congenital muscular dystrophy with secondary laminin alpha2
deficiency and abnormal glycosylation of alpha-dystroglycan. Am J Hum Genet. 69:1198, 2001a.
PMID 11592034
68. Brockington M, Torelli S, Prandini P, Boito C, Dolatshad NF, Longman C, Brown SC, Muntoni F:
Localization and functional analysis of the LARGE family of glycosyltransferases Significance for muscular
dystrophy. Hum Mol Genet. 14:657, 2005. PMID 15661757

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 49 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

69. Brockington M, Yuva Y, Prandini P, Brown SC, Torelli S, Benson MA, Herrmann R, Anderson LV,
Bashir R, Burgunder JM, Fallet S, Romero N, Fardeau M, Straub V, Storey G, Pollitt C, Richard I, Sewry
CA, Bushby K, Voit T, Blake DJ, Muntoni F: Mutations in the fukutin-related protein gene (FKRP) identify
limb girdle muscular dystrophy 2I as a milder allelic variant of congenital muscular dystrophy MDC1C.
Hum Mol Genet. 10:2851, 2001b. PMID 11741828
70. Brooke MH, Fenichel GM, Griggs RC, Mendell JR, Moxley R, Florence J, King WM, Pandya S,
Robison J, Schierbecker J: , et al Duchenne muscular dystrophy. : 475, 1989. PMID 2927672
71. Brooke MH, Fenichel GM, Griggs RC, Mendell JR, Moxley R, Miller JP, Province MA: Clinical
investigation in Duchenne dystrophy 2. Determination of the "power" of therapeutic trials based on the
natural history. Muscle Nerve. 6:91, 1983. PMID 6343858
72. Brooke MH, Fenichel GM, Griggs RC, Mendell JR, Moxley RT 3rd, Miller JP, Kaiser KK, Florence JM,
Pandya S, Signore L: , et al Clinical investigation of Duchenne muscular dystrophy. Interesting results in a
trial of prednisone. Arch Neurol. 44:812, 1987. PMID 3632393
73. Brown CS, Thomas NS, Sarfarazi M, Davies KE, Kunkel L, Pearson PL, Kingston HM, Shaw DJ,
Harper PS: Genetic linkage relationships of seven DNA probes with Duchenne and Becker muscular
dystrophy. Hum Genet. 71:62, 1985. PMID 2993158
74. Brunet G, Tome FM, Samson F, Robert JM, Fardeau M: [Oculopharyngeal muscular dystrophy. A
census of French families and genealogic study. ] Rev Neurol (Paris). 146:425, 1990. PMID 2204987
75. Buckle VJ, Guenet JL, Simon-Chazottes D, Love DR, Davies KE: Localisation of a dystrophin-related
autosomal gene to 6q24 in man, and to mouse chromosome 10 in the region of the dystrophia muscularis
(dy) locus. Hum Genet. 85:324, 1990. PMID 2203673
76. Bulfield G, Siller WG, Wight PA, Moore KJ: X chromosome-linked muscular dystrophy (mdx) in the
mouse. Proc Natl Acad Sci USA. 81:1189, 1984. PMID 6583703
77. Bullock S, Felix C, Iskander-Gabra S, Davison V: Detection of germinal mosaicism in a DMD family.
Biochem Soc Trans. 24:273S, 1996. PMID 8736931
78. Burghes AH, Logan C, Hu X, Belfall B, Worton RG, Ray PN: A cDNA clone from the
Duchenne/Becker muscular dystrophy gene. Nature X328X. : 434, 1987. PMID 3614347
79. Burkin DJ, Kaufman SJ: The alpha7beta1 integrin in muscle development and disease. Cell Tissue
Res. 296:183, 1999. PMID 10199978
80. Burn J, Povey S, Boyd Y, Munro EA, West L, Harper K, Thomas D: Duchenne muscular dystrophy in
one of monozygotic twin girls. J Med Genet. 23:494, 1986. PMID 2879922
81. Burrow KL, Coovert DD, Klein CJ, Bulman DE, Kissel JT, Rammohan KW, Burghes AH, Mendell JR:
Dystrophin expression and somatic reversion in prednisone-treated and untreated Duchenne dystrophy.
CIDD Study Group. Neurology. 41:661, 1991. PMID 1781820
82. Bushby K, Muntoni F, Bourke JP: 107th ENMC international workshop The management of cardiac
involvement in muscular dystrophy and myotonic dystrophy. 7th-9th June 2002, Naarden, the
Netherlands. Neuromuscul Disord. 13:166, 2003. PMID 12565916
83. Buzzini SR, Macmilan ACBC: gamma-Sarcoglycanopathy in two Palestinian-American siblings. Clin
Pediatr (Phila). 40:115, 2001. PMID 11261449
84. Byers TJ, Kunkel LM, Watkins SC: The subcellular distribution of dystrophin in mouse skeletal,
cardiac, and smooth muscle. J Cell Biol. 115:411, 1991. PMID 1918148
85. Byers TJ, Lidov HG, Kunkel LM: An alternative dystrophin transcript specific to peripheral nerve. Nat
Genet. 4:77, 1993. PMID 8513330
86. Cacurri S, Piazzo N, Deidda G, Vigneti E, Galluzzi G, Colantoni L, Merico B, Ricci E, Felicetti L:
Sequence homology between 4qter and 10qter loci facilitates the instability of subtelomeric KpnI repeat
units implicated in facioscapulohumeral muscular dystrophy. Am J Hum Genet. 63:181, 1998. PMID
9634507
87. Calvano S, Memeo E, Piemontese MR, Melchionda S, Bisceglia L, Gasparini P, Zelante L: Detection
of dystrophin deletion carriers using FISH analysis. Clin Genet. 52:17, 1997R. PMID 9272708
88. Camacho Vanegas O, Bertini E, Zhang RZ, Petrini S, Minosse C, Sabatelli P, Giusti B, Chu ML, Pepe

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 50 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

G: Ullrich scleroatonic muscular dystrophy is caused by recessive mutations in collagen type VI. Proc
Natl Acad Sci USA. 98:7516, 2001. PMID 11381124
89. Campbell KP, Kahl SD: Association of dystrophin and an integral membrane glycoprotein. Nature.
338:259, 1989. PMID 2493582
90. Carbone I, Bruno C, Sotgia F, Bado M, Broda P, Masetti E, Panella A, Zara F, Bricarelli FD, Cordone
G, Lisanti MP, Minetti ABC, LASTNAME ABC, LASTNAME C: Mutation in the CAV3 gene causes partial
caveolin-3 deficiency and hyperCKemia. Neurology. 54:1373, 2000. PMID 10746614
91. Carpenter S, Karpati G: Segmental necrosis and its demarcation in experimental micropuncture injury
of skeletal muscle fibers. J Neuropathol Exp Neurol. 48:154, 1989. PMID 2646398
92. Carrie A, Piccolo F, Leturcq F, de Toma C, Azibi K, Beldjord C, Vallat JM, Merlini L, Voit T, Sewry C,
Urtizberea JA, Romero N, Tome FM, Fardeau M, Sunada Y, Campbell KP, Kaplan JC, Jeanpierre M:
Mutational diversity and hot spots in the alpha-sarcoglycan gene in autosomal recessive muscular
dystrophy (LGMD2D). J Med Genet. 34:470, 1997. PMID 9192266
93. Chamberlain JS, Farwell NJ, Chamberlain JR, Cox GA, Caskey CT: PCR analysis of dystrophin gene
mutation and expression. J Cell Biochem. 46:255, 1991. PMID 1774226
94. Chamberlain JS, Gibbs RA, Ranier JE, Nguyen PN, Caskey CT: Deletion screening of the Duchenne
muscular dystrophy locus via multiplex DNA amplification. Nucleic Acids Res. 16:11141, 1988a.
PMID 3205741
95. Chamberlain JS, Pearlman JA, Muzny DM, Gibbs RA, Ranier JE, Caskey CT, Reeves AA:
Expression of the murine Duchenne muscular dystrophy gene in muscle and brain. Science.
239:1416, 1988b. PMID 3347839
96. Chambers SP, Dodd A, Overall R, Sirey T, Lam LT, Morris GE, Love DR: Dystrophin in adult
zebrafish muscle. Biochem Biophys Res Commun. 286:478, 2001. PMID 11511083
97. Chan Y, Kunkel LM: In vitro expressed dystrophin fragments do not associate with each other. FEBS
Lett. 410:153, 1997. PMID 9237620
98. Chao DS, Gorospe JR, Brenman JE, Rafael JA, Peters MF, Froehner SC, Hoffman EP, Chamberlain
JS, Bredt DS: Selective loss of sarcolemmal nitric oxide synthase in Becker muscular dystrophy. J
Exp Med. 184:609, 1996. PMID 8760814
99. Chapman VM, Miller DR, Armstrong D, Caskey CT: Recovery of induced mutations for X
chromosome-linked muscular dystrophy in mice. Proc Natl Acad Sci USA. 86:1292, 1989. PMID
2919177
100. Clarke DL, Johansson CB, Wilbertz J, Veress B, Nilsson E, Karlstrom H, Lendahl U, Frisen J:
Generalized potential of adult neural stem cells. Science. 288:1660, 2000. PMID 10834848
101. Clemens PR, Krause TL, Chan S, Korb KE, Graham FL, Caskey CT: Recombinant truncated
dystrophin minigenes Construction, expression, and adenoviral delivery. Hum Gene Ther. 6:1477,
1995. PMID 8573620
102. Clements L, Manilal S, Love DR, Morris GE: Direct interaction between emerin and lamin A. Biochem
Biophys Res Commun. 267:709, 2000. PMID 10673356
103. Coffey AJ, Roberts RG, Green ED, Cole CG, Butler R, Anand R, Giannelli F, Bentley DR:
Construction of a 2.6-Mb contig in yeast artificial chromosomes spanning the human dystrophin gene
using an STS-based approach. Genomics. 12:474, 1992. PMID 1559699
104. Cooper BJ, Winand NJ, Stedman H, Valentine BA, Hoffman EP, Kunkel LM, Scott MO, Fischbeck
KH, Kornegay JN, Avery RJ: , et al The homologue of the Duchenne locus is defective in X-linked
muscular dystrophy of dogs. Nature. 334:154, 1988. PMID 3290691
105. Coquet M, Vital C, Julien J: Presence of inclusion body myositis-like filaments in oculopharyngeal
muscular dystrophy. Ultrastructural study of 10 cases. Neuropathol Appl Neurobiol. 16:393, 1990.
PMID 2175847
106. Coral-Vazquez R, Cohn RD, Moore SA, Hill JA, Weiss RM, Davisson RL, Straub V, Barresi R, Bansal
D, Hrstka RF, Williamson R, Campbell KP: Disruption of the sarcoglycan-sarcospan complex in vascular
smooth muscle A novel mechanism for cardiomyopathy and muscular dystrophy. Cell. 98:465, 1999.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 51 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

PMID 10481911
107. Corrado K, Mills PL, Chamberlain AJSBC: Deletion analysis of the dystrophin-actin binding domain.
FEBS Lett. 344:255, 1994. PMID 8187894
108. Corrado K, Rafael JA, Mills PL, Cole NM, Faulkner JA, Wang K, Chamberlain JS: Transgenic mdx
mice expressing dystrophin with a deletion in the actin-binding domain display a "mild Becker" phenotype.
J Cell Biol. 134:873, 1996. PMID 8769413
109. Cossu G: Unorthodox myogenesis Possible developmental significance and implications for tissue
histogenesis and regeneration. Histol Histopathol. 12:755, 1997. PMID 9225158
110. Cox GA, Cole NM, Matsumura K, Phelps SF, Hauschka SD, Campbell KP, Faulkner JA, Chamberlain
JS: Overexpression of dystrophin in transgenic mdx mice eliminates dystrophic symptoms without toxicity.
Nature. 364:725, 1993. PMID 8355788
111. Cox GA, Sunada Y, Campbell KP, Chamberlain JS: Nat Genet. 8:333, 1994. PMID 7894482
112. Crawford GE, Faulkner JA, Crosbie RH, Campbell KP, Froehner SC, Chamberlain JS: Assembly of
the dystrophin-associated protein complex does not require the dystrophin COOH-terminal domain. J
Cell Biol. 150:1399, 2000. PMID 10995444
113. Crosbie RH, Heighway J, Venzke DP, Lee JC, Campbell KP: Sarcospan, the 25-kDa transmembrane
component of the dystrophin-glycoprotein complex. J Biol Chem. 272:31221, 1997. PMID 9395445
114. Crosbie RH, Lebakken CS, Holt KH, Venzke DP, Straub V, Lee JC, Grady RM, Chamberlain JS,
Sanes JR, Campbell KP: Membrane targeting and stabilization of sarcospan is mediated by the
sarcoglycan subcomplex. J Cell Biol. 145:153, 1999. PMID 10189375
115. Crosbie RH, Lim LE, Moore SA, Hirano M, Hays AP, Maybaum SW, Collin H, Dovico SA, Stolle CA,
Fardeau M, Tome FM, Campbell KP: Molecular and genetic characterization of sarcospan Insights into
sarcoglycan-sarcospan interactions. Hum Mol Genet. 9:2019, 2000. PMID 10942431
116. Cullen MJ, Jaros E: Ultrastructure of the skeletal muscle in the X chromosome-linked dystrophic
(mdx) mouse. Comparison with Duchenne muscular dystrophy. Acta Neuropathol (Berl). 77:69, 1988.
PMID 3239377
117. Cullen MJ, Walsh J, Nicholson LV, Harris JB, Zubrzycka-Gaarn EE, Ray PN, Worton RG:
Immunogold labelling of dystrophin in human muscle, using an antibody to the last 17 amino acids of the
C-terminus. Neuromuscul Disord. 1:113, 1991. PMID 1822781
118. DSouza VN, Nguyen TM, Morris GE, Karges W, Pillers DA, Ray APNBC: A novel dystrophin isoform
is required for normal retinal electrophysiology. Hum Mol Genet. 4:837, 1995. PMID 7633443
119. Dalkilic I, Kunkel LM: Muscular dystrophies Genes to pathogenesis. Curr Opin Genet Dev. 13:231,
2003. PMID 12787784
120. Davies KE, Pearson PL, Harper PS, Murray JM, OBrien T, Sarfarazi M, Williamson R: Linkage
analysis of two cloned DNA sequences flanking the Duchenne muscular dystrophy locus on the short arm
of the human X chromosome. Nucleic Acids Res. 11:2303, 1983. PMID 6304647
121. Davies KE, Young BD, Elles RG, Hill ME, Williamson R: Cloning of a representative genomic library
of the human X chromosome after sorting by flow cytometry. Nature. 293:374, 1981. PMID 6456416
122. De Sandre-Giovannoli A, Bernard R, Cau P, Navarro C, Amiel J, Boccaccio I, Lyonnet S, Stewart CL,
Munnich A, Le Merrer M, Levy N: Lamin a truncation in Hutchinson-Gilford progeria. Science.
300:2055, 2003. PMID 12702809
123. Deconinck AE, Potter AC, Tinsley JM, Wood SJ, Vater R, Young C, Metzinger L, Vincent A, Slater
CR, Davies KE: Postsynaptic abnormalities at the neuromuscular junctions of utrophin-deficient mice.
J Cell Biol. 136:883, 1997a. PMID 9049253
124. Deconinck AE, Rafael JA, Skinner JA, Brown SC, Potter AC, Metzinger L, Watt DJ, Dickson JG,
Tinsley JM, Davies KE: Utrophin-dystrophin-deficient mice as a model for Duchenne muscular dystrophy.
Cell. 90:717, 1997b. PMID 9288751
125. Deconinck N, Rafael JA, Beckers-Bleukx G, Kahn D, Deconinck AE, Davies KE, Gillis JM:
Consequences of the combined deficiency in dystrophin and utrophin on the mechanical properties and
myosin composition of some limb and respiratory muscles of the mouse. Neuromuscul Disord. 8:362,

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 52 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

1998. PMID 9713852


126. Deconinck N, Tinsley J, De Backer F, Fisher R, Kahn D, Phelps S, Davies K, Gillis JM: Expression of
truncated utrophin leads to major functional improvements in dystrophin-deficient muscles of mice.
Nat Med. 3:1216, 1997. PMID 9359695
127. Deidda G, Cacurri S, Piazzo N, Felicetti L: Direct detection of 4q35 rearrangements implicated in
facioscapulohumeral muscular dystrophy (FSHD). J Med Genet. 33:361, 1996. PMID 8733043
128. Demir E, Sabatelli P, Allamand V, Ferreiro A, Moghadaszadeh B, Makrelouf M, Topaloglu H,
Echenne B, Merlini L, Guicheney P: Mutations in COL6A3 cause severe and mild phenotypes of Ullrich
congenital muscular dystrophy. Am J Hum Genet. 70:1446, 2002. PMID 11992252
129. DeSilva S, Drachman DB, Mellits D, Kuncl RW: Prednisone treatment in Duchenne muscular
dystrophy. Long-term benefit. Arch Neurol. 44:818, 1987. PMID 3632394
130. Dincer P, Balci B, Yuva Y, Talim B, Brockington M, Dincel D, Torelli S, Brown S, Kale G, Haliloglu G,
Gerceker FO, Atalay RC, Yakicier C, Longman C, Muntoni F, Topaloglu H: A novel form of recessive limb
girdle muscular dystrophy with mental retardation and abnormal expression of alpha-dystroglycan.
Neuromuscul Disord. 13:771, 2003. PMID 14678799
131. Dincer P, Leturcq F, Richard I, Piccolo F, Yalnizoglu D, de Toma C, Akcoren Z, Broux O, Deburgrave
N, Brenguier L, Rodat C, Urtizberea JA, Jung D, Tan E, Jeanpierre M, Campbell KP, Kaplan JC,
Beckmann JS, Topaloglu H: A biochemical, genetic, and clinical survey of autosomal recessive limb girdle
muscular dystrophies in Turkey. Ann Neurol. 42:222, 1997. PMID 9266733
132. Dominski Z, Kole R: Restoration of correct splicing in thalassemic pre-mRNA by antisense
oligonucleotides. Proc Natl Acad Sci USA. 90:8673, 1993. PMID 8378346
133. Drachman DB, Toyka KV, Myer E: Prednisone in Duchenne muscular dystrophy. Lancet. 2:1409,
1974. PMID 4140328
134. Dreyfus JC, Schapira G, Demos J: [Study of serum creatine kinase in myopathic patients and their
families. ] Rev Fr Etud Clin Biol. 5:384, 1960. PMID 13818100
135. Duchenne GBA: Paraplegie cerebrale, congenitale, hypertrophique, in LElectrisation Localisee et de
son Application a la Pathologie et a la Therapeutique. Paris, J-B Bailliere et Fils, 1861
136. Duchenne GBA: Recherches sur la paralysie musculaire pseudo-hypertrophique ou paralysie
myosclerosique. Arch Gen Med. 11:5, 178, 305, 421, 552, 1868.
137. Duclos F, Straub V, Moore SA, Venzke DP, Hrstka RF, Crosbie RH, Durbeej M, Lebakken CS,
Ettinger AJ, van der Meulen J, Holt KH, Lim LE, Sanes JR, Davidson BL, Faulkner JA, Williamson R,
Campbell KP: Progressive muscular dystrophy in alpha-sarcoglycan-deficient mice. J Cell Biol.
142:1461, 1998. PMID 9744877
138. Duggan DJ, Fanin M, Pegoraro E, Angelini C, Hoffman EP: alpha-Sarcoglycan (adhalin) deficiency
Complete deficiency patients are 5% of childhood-onset dystrophin-normal muscular dystrophy and most
partial deficiency patients do not have gene mutations. J Neurol Sci. 140:30, 1996. PMID 8866424
139. Duggan DJ, Gorospe JR, Fanin M, Hoffman EP, Angelini C: Mutations in the sarcoglycan genes in
patients with myopathy. N Engl J Med. 336:618, 1997a. PMID 9032047
140. Duggan DJ, Manchester D, Stears KP, Mathews DJ, Hart C, Hoffman EP: Mutations in the
delta-sarcoglycan gene are a rare cause of autosomal recessive limb-girdle muscular dystrophy (LGMD2).
Neurogenetics. 1:49, 1997b. PMID 10735275
141. Durbeej M, Campbell KP: Muscular dystrophies involving the dystrophin-glycoprotein complex An
overview of current mouse models. Curr Opin Genet Dev. 12:349, 2002. PMID 12076680
142. Eagle M, Baudouin SV, Chandler C, Giddings DR, Bullock R, Bushby K: Survival in Duchenne
muscular dystrophy Improvements in life expectancy since 1967 and the impact of home nocturnal
ventilation. Neuromuscul Disord. 12:926, 2002. PMID 12467747
143. Ebashi S, Toyokura Y, Momoi H, Sugita H: High creatine phosphokinase activity of sera of
progressive muscular dystrophy. J Biochem. 46:103, 1959.
144. Emery AEH, Emery MLH: The History of a Genetic Disease. : Duchenne Muscular Dystrophy or
Meryons Disease. London, The Royal Society of Medicine Press, 1995.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 53 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

145. English KM, Gibbs JL: Cardiac monitoring and treatment for children and adolescents with
neuromuscular disorders. Dev Med Child Neurol. 48:231, 2006. PMID 16483403
146. Eriksson M, Brown WT, Gordon LB, Glynn MW, Singer J, Scott L, Erdos MR, Robbins CM, Moses
TY, Berglund P, Dutra A, Pak E, Durkin S, Csoka AB, Boehnke M, Glover TW, Collins FS: Recurrent de
novo point mutations in lamin A cause Hutchinson-Gilford progeria syndrome. Nature. 423:293, 2003.
PMID 12714972
147. Ervasti JM, Campbell KP: Membrane organization of the dystrophin-glycoprotein complex. Cell.
66:1121, 1991. PMID 1913804
148. Ervasti JM, Ohlendieck K, Kahl SD, Gaver MG, Campbell KP: Deficiency of a glycoprotein component
of the dystrophin complex in dystrophic muscle. Nature. 345:315, 1990. PMID 2188135
149. Ettinger AJ, Feng G, Sanes JR: epsilon-Sarcoglycan, a broadly expressed homologue of the gene
mutated in limb-girdle muscular dystrophy 2D. J Biol Chem. 272:32534, 1997. PMID 9405466
150. Eymard B, Romero NB, Leturcq F, Piccolo F, Carrie A, Jeanpierre M, Collin H, Deburgrave N, Azibi
K, Chaouch M, Merlini L, Themar-Noel C, Penisson I, Mayer M, Tanguy O, Campbell KP, Kaplan JC,
Tome FM, Fardeau M: Primary adhalinopathy (alpha-sarcoglycanopathy) Clinical, pathologic, and genetic
correlation in 20 patients with autosomal recessive muscular dystrophy. Neurology. 48:1227, 1997.
PMID 9153448
151. Fadic R, Sunada Y, Waclawik AJ, Buck S, Lewandoski PJ, Campbell KP, Lotz BP: Brief report
Deficiency of a dystrophin-associated glycoprotein (adhalin) in a patient with muscular dystrophy and
cardiomyopathy. N Engl J Med. 334:362, 1996. PMID 8538707
152. Farrell PT: Anaesthesia-induced rhabdomyolysis causing cardiac arrest Case report and review of
anaesthesia and the dystrophinopathies. Anaesth Intensive Care. 22:597, 1994. PMID 7818067
153. Feener CA, Koenig M, Kunkel LM: Alternative splicing of human dystrophin mRNA generates
isoforms at the carboxy terminus. Nature. 338:509, 1989. PMID 2648158
154. Feit H, Silbergleit A, Schneider LB, Gutierrez JA, Fitoussi RP, Reyes C, Rouleau GA, Brais B,
Jackson CE, Beckmann JS, Seboun E: Vocal cord and pharyngeal weakness with autosomal dominant
distal myopathy Clinical description and gene localization to 5q31. Am J Hum Genet. 63:1732, 1998.
PMID 9837826
155. Fenichel GM, Florence JM, Pestronk A, Mendell JR, Moxley RT 3rd, Griggs RC, Brooke MH, Miller
JP, Robison J, King W: , et al Long-term benefit from prednisone therapy in Duchenne muscular
dystrophy. Neurology. 41:1874, 1991a. PMID 1745340
156. Fenichel GM, Mendell JR, Moxley RT 3rd, Griggs RC, Brooke MH, Miller JP, Pestronk A, Robison J,
King W, Signore L: A comparison of daily and alternate-day prednisone therapy in the treatment of
Duchenne muscular dystrophy. Arch Neurol. 48:575, 1991b. PMID 2039377
157. Ferlini A, Galie N, Merlini L, Sewry C, Branzi A, Muntoni F: A novel Alu-like element rearranged in the
dystrophin gene causes a splicing mutation in a family with X-linked dilated cardiomyopathy. Am J
Hum Genet. 63:436, 1998. PMID 9683584
158. Ferrari G, Cusella-De Angelis G, Coletta M, Paolucci E, Stornaiuolo A, Cossu G, Mavilio F: Muscle
regeneration by bone marrow-derived myogenic progenitors. Science. 279:1528, 1998. PMID
9488650
159. Flanigan KM, von Niederhausern A, Dunn DM, Alder J, Mendell JR, Weiss RB: Rapid direct
sequence analysis of the dystrophin gene. Am J Hum Genet. 72:931, 2003. PMID 12632325
160. Forood B, Perez-Paya E, Houghten RA, Blondelle SE: Formation of an extremely stable polyalanine
beta-sheet macromolecule. Biochem Biophys Res Commun. 211:7, 1995. PMID 7779112
161. Francke U, Ochs HD, de Martinville B, Giacalone J, Lindgren V, Disteche C, Pagon RA, Hofker MH,
van Ommen GJ, Pearson PL: , et al Minor Xp21 chromosome deletion in a male associated with
expression of Duchenne muscular dystrophy, chronic granulomatous disease, retinitis pigmentosa, and
McLeod syndrome. Am J Hum Genet. 37:250, 1985. PMID 4039107
162. Fried K, Arlozorov A, Spira R: Autosomal recessive oculopharyngeal muscular dystrophy. J Med
Genet. 12:416, 1975. PMID 1219124

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 54 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

163. 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. Am J Hum Genet. 70:663, 2002. PMID 11822024
164. Fukuyama Y, Osawa M, Suzuki H: Congenital progressive muscular dystrophy of the Fukuyama type
- clinical, genetic and pathological considerations. Brain Dev. 3:1, 1981. PMID 7258547
165. Funakoshi M, Goto K, Arahata K: Epilepsy and mental retardation in a subset of early onset
4q35-facioscapulohumeral muscular dystrophy. Neurology. 50:1791, 1998. PMID 9633729
166. Gabellini D, DAntona G, Moggio M, Prelle A, Zecca C, Adami R, Angeletti B, Ciscato P, Pellegrino
MA, Bottinelli R, Green MR, Tupler R: Facioscapulohumeral muscular dystrophy in mice overexpressing
FRG1. Nature. 439:973, 2006. PMID 16341202
167. Gabellini D, Green MR, Tupler R: Inappropriate gene activation in FSHD A repressor complex binds a
chromosomal repeat deleted in dystrophic muscle. Cell. 110:339, 2002. PMID 12176321
168. Galbiati F, Volonte D, Chu JB, Li M, Fine SW, Fu M, Bermudez J, Pedemonte M, Weidenheim KM,
Pestell RG, Minetti C, Lisanti MP: Transgenic overexpression of caveolin-3 in skeletal muscle fibers
induces a Duchenne-like muscular dystrophy phenotype. Proc Natl Acad Sci USA. 97:9689, 2000.
PMID 10931944
169. Galvagni F, Saad FA, Danieli GA, Miorin M, Vitiello L, Mostacciuolo ML, Angelini C: A study on
duplications of the dystrophin gene Evidence of a geographical difference in the distribution of breakpoints
by intron. Hum Genet. 94:83, 1994. PMID 8034300
170. Gillard EF, Chamberlain JS, Murphy EG, Duff CL, Smith B, Burghes AH, Thompson MW, Sutherland
J, Oss I, Bodrug ABC: , et al Molecular and phenotypic analysis of patients with deletions within the
deletion-rich region of the Duchenne muscular dystrophy (DMD) gene. Am J Hum Genet. 45:507,
1989. PMID 2491010
171. Ginjaar HB, van der Kooi AJ, Ceelie H, Kneppers AL, van Meegen M, Barth PG, Busch HF, Wokke
JH, Anderson LV, Bonnemann CG, Jeanpierre M, Bolhuis PA, Moorman AF, de Visser M, Bakker E,
Ommen GJ: Sarcoglycanopathies in Dutch patients with autosomal recessive limb girdle muscular
dystrophy. J Neurol. 247:524, 2000. PMID 10993494
172. Gnecchi-Ruscone T, Taylor J, Mercuri E, Paternostro G, Pogue R, Bushby K, Sewry C, Muntoni F,
Camici PG: Cardiomyopathy in duchenne, becker, and sarcoglycanopathies A role for coronary
dysfunction? Muscle Nerve. 22:1549, 1999. PMID 10514233
173. Goodell MA, Brose K, Paradis G, Conner AS, Mulligan RC: Isolation and functional properties of
murine hematopoietic stem cells that are replicating in vivo. J Exp Med. 183:1797, 1996. PMID
8666936
174. Gowers WR: Pseudo-Hypertrophic Muscular Paralysis--A Clinical Lecture. London, J & A Churchill,
1879
175. Grady RM, Grange RW, Lau KS, Maimone MM, Nichol MC, Stull JT, Sanes JR: Role for
alpha-dystrobrevin in the pathogenesis of dystrophin-dependent muscular dystrophies. Nat Cell Biol.
1:215, 1999. PMID 10559919
176. Grady RM, Merlie JP, Sanes JR: Subtle neuromuscular defects in utrophin-deficient mice. J Cell Biol.
136:871, 1997a. PMID 9049252
177. Grady RM, Teng H, Nichol MC, Cunningham JC, Wilkinson RS, Sanes JR: Skeletal and cardiac
myopathies in mice lacking utrophin and dystrophin A model for Duchenne muscular dystrophy. Cell.
90:729, 1997b. PMID 9288752
178. Grady RM, Wozniak DF, Ohlemiller KK, Sanes JR: Cerebellar synaptic defects and abnormal motor
behavior in mice lacking alpha- and beta-dystrobrevin. J Neurosci. 26:2841, 2006. PMID 16540561
179. Graham FL, Smiley J, Russell WC, Nairn R: Characteristics of a human cell line transformed by DNA
from human adenovirus type 5. J Gen Virol. 36:59, 1977. PMID 886304
180. Granato M, van Eeden FJ, Schach U, Trowe T, Brand M, Furutani-Seiki M, Haffter P,
Hammerschmidt M, Heisenberg CP, Jiang YJ, Kane DA, Kelsh RN, Mullins MC, Odenthal J,
Nusslein-Volhard C: Genes controlling and mediating locomotion behavior of the zebrafish embryo and

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 55 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

larva. Development. 123:399, 1996. PMID 9007258


181. Gregorevic P, Blankinship MJ, Allen JM, Crawford RW, Meuse L, Miller DG, Russell DW,
Chamberlain JS: Systemic delivery of genes to striated muscles using adeno-associated viral vectors.
Nat Med. 10:828, 2004. PMID 15273747
182. Grewal PK, Holzfeind PJ, Bittner RE, Hewitt JE: Mutant glycosyltransferase and altered glycosylation
of alpha-dystroglycan in the myodystrophy mouse. Nat Genet. 28:151, 2001. PMID 11381262
183. Grifo JA, Tang YX, Munne S, Alikani M, Cohen J, Rosenwaks Z: Healthy deliveries from biopsied
human embryos. Hum Reprod. 9:912, 1994. PMID 7929742
184. Griggs RC, Bushby K: Continued need for caution in the diagnosis of Duchenne muscular dystrophy.
Neurology. 64:1498, 2005. PMID 15883307
185. Griggs RC, Moxley RT 3rd, Mendell JR, Fenichel GM, Brooke MH, Pestronk A, Miller JP: Prednisone
in Duchenne dystrophy. A randomized, controlled trial defining the time course and dose response.
Clinical Investigation of Duchenne Dystrophy Group. Arch Neurol. 48:383, 1991. PMID 2012511
186. Griggs RC, Moxley RT 3rd, Mendell JR, Fenichel GM, Brooke MH, Pestronk A, Miller JP, Cwik VA,
Pandya S, Robison J: , et al Duchenne dystrophy. : 520, 1993. PMID 8450994
187. Grimm T, Meng G, Liechti-Gallati S, Bettecken T, Mller CR, Mller 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. J Med Genet. 31:183, 1994. PMID 8014964
188. Grobet L, Martin LJ, Poncelet D, Pirottin D, Brouwers B, Riquet J, Schoeberlein A, Dunner S,
Menissier F, Massabanda J, Fries R, Hanset R, Georges M: A deletion in the bovine myostatin gene
causes the double-muscled phenotype in cattle. Nat Genet. 17:71, 1997. PMID 9288100
189. Grobet L, Poncelet D, Royo LJ, Brouwers B, Pirottin D, Michaux C, Menissier F, Zanotti M, Dunner S,
Georges M: Molecular definition of an allelic series of mutations disrupting the myostatin function and
causing double-muscling in cattle. Mamm Genome. 9:210, 1998. PMID 9501304
190. Guicheney P, Vignier N, Zhang X, He Y, Cruaud C, Frey V, Helbling-Leclerc A, Richard P, Estournet
B, Merlini L, Topaloglu H, Mora M, Harpey JP, Haenggeli CA, Barois A, Hainque B, Schwartz K, Tome
FM, Fardeau M, Tryggvason K: PCR based mutation screening of the laminin alpha2 chain gene (LAMA2)
Application to prenatal diagnosis and search for founder effects in congenital muscular dystrophy. J
Med Genet. 35:211, 1998. PMID 9541105
191. 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. J Neurol Neurosurg Psychiatry. 65:225, 1998. PMID 9703177
192. Gussoni E, Blau HM, Kunkel LM: The fate of individual myoblasts after transplantation into muscles of
DMD patients. Nat Med. 3:970, 1997. PMID 9288722
193. Gussoni E, Pavlath GK, Lanctot AM, Sharma KR, Miller RG, Steinman L, Blau HM: Normal dystrophin
transcripts detected in Duchenne muscular dystrophy patients after myoblast transplantation. Nature.
356:435, 1992. PMID 1557125
194. Gussoni E, Soneoka Y, Strickland CD, Buzney EA, Khan MK, Flint AF, Kunkel LM, Mulligan RC:
Dystrophin expression in the mdx mouse restored by stem cell transplantation. Nature. 401:390,
1999. PMID 10517639
195. Gussoni E, Wang Y, Fraefel C, Miller RG, Blau HM, Geller AI, Kunkel LM: A method to codetect
introduced genes and their products in gene therapy protocols. Nat Biotechnol. 14:1012, 1996. PMID
9631042
196. Guyon JR, Kudryashova E, Potts A, Dalkilic I, Brosius MA, Thompson TG, Beckmann JS, Kunkel LM,
Spencer MJ: Calpain 3 cleaves filamin C and regulates its ability to interact with gamma- and
delta-sarcoglycans. Muscle Nerve. 28:472, 2003a. PMID 14506720
197. Guyon JR, Mosley AN, Jun SJ, Montanaro F, Steffen LS, Zhou Y, Nigro V, Zon LI, Kunkel LM:
Delta-sarcoglycan is required for early zebrafish muscle organization. Exp Cell Res. 304:105, 2005.
PMID 15707578
198. Guyon JR, Mosley AN, Zhou Y, OBrien KF, Sheng X, Chiang K, Davidson AJ, Volinski JM, Zon LI,

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 56 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Kunkel LM: The dystrophin associated protein complex in zebrafish. Hum Mol Genet. 12:601, 2003b.
PMID 12620966
199. Hack AA, Ly CT, Jiang F, Clendenin CJ, Sigrist KS, Wollmann RL, McNally EM: Gamma-sarcoglycan
deficiency leads to muscle membrane defects and apoptosis independent of dystrophin. J Cell Biol.
142:1279, 1998. PMID 9732288
200. Hackman P, Vihola A, Haravuori H, Marchand S, Sarparanta J, De Seze J, Labeit S, Witt C, Peltonen
L, Richard I, Udd B: Tibial muscular dystrophy is a titinopathy caused by mutations in TTN, the gene
encoding the giant skeletal-muscle protein titin. Am J Hum Genet. 71:492, 2002. PMID 12145747
201. Haffter P, Granato M, Brand M, Mullins MC, Hammerschmidt M, Kane DA, Odenthal J, van Eeden FJ,
Jiang YJ, Heisenberg CP, Kelsh RN, Furutani-Seiki M, Vogelsang E, Beuchle D, Schach U, Fabian C,
Nusslein-Volhard C: The identification of genes with unique and essential functions in the development of
the zebrafish, Danio rerio. Development. 123:1, 1996.. PMID 9007226
202. Haravuori H, Vihola A, Straub V, Auranen M, Richard I, Marchand S, Voit T, Labeit S, Somer H,
Peltonen L, Beckmann JS, Udd B: Secondary calpain3 deficiency in 2q-linked muscular dystrophy Titin is
the candidate gene. Neurology. 56:869, 2001. PMID 11294923
203. Haslett JN, Kang PB, Han M, Kho AT, Sanoudou D, Volinski JM, Beggs AH, Kohane IS, Kunkel LM:
The influence of muscle type and dystrophin deficiency on murine expression profiles. Mamm
Genome. 16:739, 2005. PMID 16261416
204. Hauser MA, Horrigan SK, Salmikangas P, Torian UM, Viles KD, Dancel R, Tim RW, Taivainen A,
Bartoloni L, Gilchrist JM, Stajich JM, Gaskell PC, Gilbert JR, Vance JM, Pericak-Vance MA, Carpn O,
Westbrook CA, Speer MC: Myotilin is mutated in limb girdle muscular dystrophy 1A. Hum Mol Genet.
9:2141, 2000. PMID 10958653
205. Hayashi YK, Chou FL, Engvall E, Ogawa M, Matsuda C, Hirabayashi S, Yokochi K, Ziober BL,
Kramer RH, Kaufman SJ, Ozawa E, Goto Y, Nonaka I, Tsukahara T, Wang JZ, Hoffman EP, Arahata K:
Mutations in the integrin alpha7 gene cause congenital myopathy. Nat Genet. 19:94, 1998. PMID
9590299
206. Helbling-Leclerc A, Zhang X, Topaloglu H, Cruaud C, Tesson F, Weissenbach J, Tome FM, Schwartz
K, Fardeau M, Tryggvason K: , et al Mutations in the laminin alpha 2-chain gene (LAMA2) cause
merosin-deficient congenital muscular dystrophy. Nat Genet. 11:216, 1995. PMID 7550355
207. Henry MD, Campbell KP: A role for dystroglycan in basement membrane assembly. Cell. 95:859,
1998. PMID 9865703
208. Hewitt JE, Lyle R, Clark LN, Valleley EM, Wright TJ, Wijmenga C, van Deutekom JC, Francis F,
Sharpe PT, Hofker M: , et al Analysis of the tandem repeat locus D4Z4 associated with
facioscapulohumeral muscular dystrophy. Hum Mol Genet. 3:1287, 1994. PMID 7987304
209. Higuchi I, Iwaki H, Kawai H, Endo T, Kunishige M, Fukunaga H, Nakagawa M, Arimura K, Osame M:
New missense mutation in the alpha-sarcoglycan gene in a Japanese patient with severe childhood
autosomal recessive muscular dystrophy with incomplete alpha-sarcoglycan deficiency. J Neurol Sci.
153:100, 1997. PMID 9455986
210. Higuchi I, Shiraishi T, Hashiguchi T, Suehara M, Niiyama T, Nakagawa M, Arimura K, Maruyama I,
Osame M: Frameshift mutation in the collagen VI gene causes Ullrichs disease. Ann Neurol. 50:261,
2001. PMID 11506412
211. Hillaire D, Leclerc A, Faure S, Topaloglu H, Chiannilkulchai N, Guicheney P, Grinas L, Legos P,
Philpot J, Evangelista T: , et al Localization of merosin-negative congenital muscular dystrophy to
chromosome 6q2 by homozygosity mapping. Hum Mol Genet. 3:1657, 1994. PMID 7833925
212. Hillier BJ, Christopherson KS, Prehoda KE, Bredt DS, Lim WA: Unexpected modes of PDZ domain
scaffolding revealed by structure of nNOS-syntrophin complex. Science. 284:812, 1999. PMID
10221915
213. Hoffman EP, Brown RH Jr, Kunkel LM: Dystrophin The protein product of the Duchenne muscular
dystrophy locus. Cell. 51:919, 1987. PMID 3319190
214. Hoffman EP, Fischbeck KH, Brown RH, Johnson M, Medori R, Loike JD, Harris JB, Waterston R,

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 57 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Brooke M, Specht L: , et al Characterization of dystrophin in muscle-biopsy specimens from patients with


Duchennes or Beckers muscular dystrophy. N Engl J Med. 318:1363, 1988a. PMID 3285207
215. Hoffman EP, Gorospe JR: The animal models of Duchenne muscular dystrophy. : Windows on the
pathophysiological consequences of dystrophin deficiency, in Ordering the Membrane Cytoskeletal
Trilayer, MT Mooseker, J Morrow (eds). New York, Academic, 1991. PMID 3078411
216. Hoffman EP, Hudecki MS, Rosenberg PA, Pollina CM, Kunkel LM: Cell and fiber-type distribution of
dystrophin. Neuron. 1:411, 1988b. PMID 3078411
217. Holt KH, Lim LE, Straub V, Venzke DP, Duclos F, Anderson RD, Davidson BL, Campbell KP:
Functional rescue of the sarcoglycan complex in the BIO 14.6 hamster using delta-sarcoglycan gene
transfer. Mol Cell. 1:841, 1998. PMID 9660967
218. Hopkins LC, Jackson JA, Elsas LJ: Emery-dreifuss humeroperoneal muscular dystrophy An x-linked
myopathy with unusual contractures and bradycardia. Ann Neurol. 10:230, 1981. PMID 7294729
219. Howard PL, Dally GY, Wong MH, Ho A, Weleber RG, Pillers DA, Ray PN: Localization of dystrophin
isoform Dp71 to the inner limiting membrane of the retina suggests a unique functional contribution of
Dp71 in the retina. Hum Mol Genet. 7:1385, 1998. PMID 9700191
220. Hu X, Bulman DE, Ray PN, Worton RG: Frameshift duplication resulting in truncated dystrophin in a
patient with Duchenne muscular dystrophy. Hum Mutat. 1:172, 1992. PMID 1301205
221. Hu XY, Burghes AH, Ray PN, Thompson MW, Murphy EG, Worton RG: Partial gene duplication in
Duchenne and Becker muscular dystrophies. J Med Genet. 25:369, 1988. PMID 3398004
222. Hu XY, Ray PN, Murphy EG, Thompson MW, Worton RG: Duplicational mutation at the Duchenne
muscular dystrophy locus Its frequency, distribution, origin, and phenotypegenotype correlation. Am J
Hum Genet. 46:682, 1990. PMID 2316519
223. Huard J, Bouchard JP, Roy R, Malouin F, Dansereau G, Labrecque C, Albert N, Richards CL,
Lemieux B, Tremblay JP: Human myoblast transplantation preliminary results of 4 cases. Muscle
Nerve. 15:550, 1992a. PMID 1584246
224. Huard J, Fortier LP, Dansereau G, Labrecque C, Tremblay JP: A light and electron microscopic study
of dystrophin localization at the mouse neuromuscular junction. Synapse. 10:83, 1992b. PMID
1585259
225. Ibraghimov-Beskrovnaya O, Ervasti JM, Leveille CJ, Slaughter CA, Sernett SW, Campbell KP:
Primary structure of dystrophin-associated glycoproteins linking dystrophin to the extracellular matrix.
Nature. 355:696, 1992. PMID 1741056
226. Ichikawa Y, Watanabe M, Kowa H, Murayama S, Mizuno ABCT, Komuro I, Ishiki R, Goto J,
Kanazawa I: A Japanese family carrying a novel mutation in the Emery-Dreifuss muscular dystrophy
gene. Ann Neurol. 41:399, 1997. PMID 9066362
227. Ishiura S, Arahata K, Tsukahara T, Koga R, Anraku H, Yamaguchi M, Kikuchi T, Nonaka I, Sugita H:
Antibody against the C-terminal portion of dystrophin crossreacts with the 400 kDa protein in the pia mater
of dystrophin-deficient mdx mouse brain. J Biochem (Tokyo). 107:510, 1990. PMID 2193022
228. Jackson KA, Mi T, Goodell MA: Hematopoietic potential of stem cells isolated from murine skeletal
muscle. Proc Natl Acad Sci USA. 96:14482, 1999. PMID 10588731
229. Jacobs SC, Bootsma AL, Willems PW, Bar PR, Wokke JH: Prednisone can protect against
exercise-induced muscle damage. J Neurol. 243:410, 1996. PMID 8741082
230. Jensen V: The anaesthetic management of a patient with Emery-Dreifuss muscular dystrophy. Can J
Anaesth. 43:968, 1996. PMID 8874917
231. Jeppesen J, Green A, Steffensen BF, Rahbek J: The Duchenne muscular dystrophy population in
Denmark, 1977-2001 Prevalence, incidence and survival in relation to the introduction of ventilator use.
Neuromuscul Disord. 13:804, 2003. PMID 14678803
232. Jobsis GJ, Boers JM, Barth PG, de Visser M: Bethlem myopathy A slowly progressive congenital
muscular dystrophy with contractures. Brain. 122:649, 1999. PMID 10219778
233. Jobsis GJ, Keizers H, Vreijling JP, de Visser M, Speer MC, Wolterman RA, Baas F, Bolhuis PA: Type
VI collagen mutations in Bethlem myopathy, an autosomal dominant myopathy with contractures. Nat

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 58 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Genet. 14:113, 1996. PMID 8782832


234. Jung D, Duclos F, Apostol B, Straub V, Lee JC, Allamand V, Venzke DP, Sunada Y, Moomaw CR,
Leveille CJ, Slaughter CA, Crawford TO, McPherson JD, Campbell KP: Characterization of
delta-sarcoglycan, a novel component of the oligomeric sarcoglycan complex involved in limb-girdle
muscular dystrophy. J Biol Chem. 271:32321, 1996. PMID 8943294
235. Jung D, Yang B, Meyer J, Chamberlain JS, Campbell KP: Identification and characterization of the
dystrophin anchoring site on beta-dystroglycan. J Biol Chem. 270:27305, 1995. PMID 7592992
236. Kambadur R, Sharma M, Smith TP, Bass JJ: Mutations in myostatin (GDF8) in double-muscled
Belgian Blue and Piedmontese cattle. Genome Res. 7:910, 1997. PMID 9314496
237. Kaminski HJ, al-Hakim M, Leigh RJ, Katirji MB, Ruff RL: Extraocular muscles are spared in advanced
Duchenne dystrophy. Ann Neurol. 32:586, 1992. PMID 1456746
238. Kamoshita S, Konishi Y, Segawa M, Fukuyama Y: Congenital muscular dystrophy as a disease of the
central nervous system. Arch Neurol. 33:513, 1976. PMID 938276
239. Karpati G, Ajdukovic D, Arnold D, Gledhill RB, Guttmann R, Holland P, Koch PA, Shoubridge E,
Spence D, Vanasse M: , et al Myoblast transfer in Duchenne muscular dystrophy. Ann Neurol. 34:8,
1993a. PMID 8517684
240. Karpati G, Carpenter S: Small-caliber skeletal muscle fibers do not suffer deleterious consequences
of dystrophic gene expression. Am J Med Genet. 25:653, 1986. PMID 3789023
241. Karpati G, Carpenter S, Morris GE, Davies KE, Guerin C, Holland P: Localization and quantitation of
the chromosome 6-encoded dystrophin-related protein in normal and pathological human muscle. J
Neuropathol Exp Neurol. 52:119, 1993b. PMID 8440993
242. Kawai H, Akaike M, Kunishige M, Inui T, Adachi K, Kimura C, Kawajiri M, Nishida Y, Endo I,
Kashiwagi S, Nishino H, Fujiwara T, Okuno S, Rodat C, Richard I, Beckmann JS, Miyoshi K,
Matsumoto T: Clinical, pathological, and genetic features of limb-girdle muscular dystrophy type 2A with
new calpain 3 gene mutations in seven patients from three Japanese families. Muscle Nerve.
21:1493, 1998. PMID 9771675
243. Kho AT, Kang PB, Kohane IS, Kunkel LM: Transcriptome-scale similarities between mouse and
human skeletal muscles with normal and myopathic phenotypes. BMC Musculoskelet Disord. 7:23,
2006. PMID 16522209
244. Khurana TS, Hoffman EP, Kunkel LM: Identification of a chromosome 6-encoded dystrophin-related
protein. J Biol Chem. 265:16717, 1990.. PMID 2170350
245. Khurana TS, Prendergast RA, Alameddine HS, Tome FM, Fardeau M, Arahata K, Sugita H, Kunkel
LM: Absence of extraocular muscle pathology in Duchennes muscular dystrophy Role for calcium
homeostasis in extraocular muscle sparing. J Exp Med. 182:467, 1995. PMID 7629506
246. Khurana TS, Watkins SC, Chafey P, Chelly J, Tome FM, Fardeau M, Kaplan JC, Kunkel LM:
Immunolocalization and developmental expression of dystrophin related protein in skeletal muscle.
Neuromuscul Disord. 1:185, 1991R. PMID 1822793
247. Kingston HM, Sarfarazi M, Thomas NS, Harper PS: Localisation of the Becker muscular dystrophy
gene on the short arm of the X chromosome by linkage to cloned DNA sequences. Hum Genet. 67:6,
1984. PMID 6086495
248. Kissel JT, Burrow KL, Rammohan KW, Mendell JR: Mononuclear cell analysis of muscle biopsies in
prednisone-treated and untreated Duchenne muscular dystrophy. CIDD Study Group. Neurology.
41:667, 1991. PMID 2027481
249. Kissel JT, Lynn DJ, Rammohan KW, Klein JP, Griggs RC, Moxley RT 3rd, Cwik VA, Brooke MH,
Mendell JR: Mononuclear cell analysis of muscle biopsies in prednisone- and azathioprine-treated
Duchenne muscular dystrophy. Neurology. 43:532, 1993. PMID 8450996
250. Kissel JT, McDermott MP, Mendell JR, King WM, Pandya S, Griggs RC, Tawil R: Randomized,
double-blind, placebo-controlled trial of albuterol in facioscapulohumeral dystrophy. Neurology.
57:1434, 2001. PMID 11673585
251. Kissel JT, McDermott MP, Natarajan R, Mendell JR, Pandya S, King WM, Griggs RC, Tawil R: Pilot

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 59 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

trial of albuterol in facioscapulohumeral muscular dystrophy. FSH-DY Group. Neurology. 50:1402,


1998. PMID 9595995
252. Klauck SM, Wilgenbus P, Yates JR, Mller CR, Poustka A: Identification of novel mutations in three
families with Emery-Dreifuss muscular dystrophy. Hum Mol Genet. 4:1853, 1995. PMID 8595406
253. Kobayashi K, Nakahori Y, Miyake M, Matsumura K, Kondo-Iida E, Nomura Y, Segawa M, Yoshioka
M, Saito K, Osawa M, Hamano K, Sakakihara Y, Nonaka I, Nakagome Y, Kanazawa I, Nakamura Y,
Tokunaga K, Toda T: An ancient retrotransposal insertion causes Fukuyama-type congenital muscular
dystrophy. Nature. 394:388, 1998a. PMID 9690476
254. Kobayashi K, Nakahori Y, Mizuno K, Miyake M, Kumagai T, Honma A, Nonaka I, Nakamura Y,
Tokunaga K, Toda TABC: Founder-haplotype analysis in Fukuyama-type congenital muscular dystrophy
(FCMD). Hum Genet. 103:323, 1998b. PMID 9799088
255. Koenig M, Beggs AH, Moyer M, Scherpf S, Heindrich K, Bettecken T, Meng G, Mller CR, Lindlof
M, Kriinen H: , et al The molecular basis for Duchenne versus Becker muscular dystrophy. :
498, 1989. PMID 2491009
256. Koenig M, Hoffman EP, Bertelson CJ, Monaco AP, Feener C, Kunkel LM: Complete cloning of the
Duchenne muscular dystrophy (DMD) cDNA and preliminary genomic organization of the DMD gene in
normal and affected individuals. Cell. 50:509, 1987. PMID 3607877
257. Koenig M, Kunkel LM: Detailed analysis of the repeat domain of dystrophin reveals four potential
hinge segments that may confer flexibility. J Biol Chem. 265:4560, 1990. PMID 2407739
258. Koenig M, Monaco AP, Kunkel LM: The complete sequence of dystrophin predicts a rod-shaped
cytoskeletal protein. Cell. 53:219, 1988. PMID 3282674
259. Kondo-Iida E, Saito K, Tanaka H, Tsuji S, Ishihara T, Osawa M, Fukuyama Y, Toda T: Molecular
genetic evidence of clinical heterogeneity in Fukuyama-type congenital muscular dystrophy. Hum
Genet. 99:427, 1997. PMID 9099829
260. Kornegay JN, Tuler SM, Miller DM, Levesque DC: Muscular dystrophy in a litter of golden retriever
dogs. Muscle Nerve. 11:1056, 1988. PMID 3185600
261. Krause S, Fakan S, Weis K, Wahle E: Immunodetection of poly(A) binding protein II in the cell
nucleus. Exp Cell Res. 214:75, 1994. PMID 8082750
262. Kristjansson K, Chong SS, Van den Veyver IB, Subramanian S, Snabes MC, Hughes MR:
Preimplantation single cell analyses of dystrophin gene deletions using whole genome amplification.
Nat Genet. 6:19, 1994. PMID 8136827
263. Kubisch C, Schoser BG, von During M, Betz RC, Goebel HH, Zahn S, Ehrbrecht A, Aasly J, Schroers
A, Popovic N, Lochmuller H, Schroder JM, Bruning T, Malin JP, Fricke B, Meinck HM, Torbergsen T,
Engels H, Voss B, Vorgerd M: Homozygous mutations in caveolin-3 cause a severe form of rippling
muscle disease. Ann Neurol. 53:512, 2003. PMID 12666119
264. Kunkel LM: Analysis of deletions in DNA from patients with Becker and Duchenne muscular
dystrophy. Nature. 322:73, 1986. PMID 3014348
265. Kunkel LM: 2004 William Allan Award address. Cloning of the DMD gene Am J Hum Genet 76 205,
2005 PMID 15714686
266. Kunkel LM, Monaco AP, Middlesworth W, Ochs HD, Latt SA: Specific cloning of DNA fragments
absent from the DNA of a male patient with an X chromosome deletion. Proc Natl Acad Sci USA.
82:4778, 1985. PMID 2991893
267. Kunkel LM, Tantravahi U, Eisenhard M, Latt SA: Regional localization on the human X of DNA
segments cloned from flow sorted chromosomes. Nucleic Acids Res. 10:1557, 1982. PMID 6461845
268. Laforet P, de Toma C, Eymard B, Becane HM, Jeanpierre M, Fardeau M, Duboc D: Cardiac
involvement in genetically confirmed facioscapulohumeral muscular dystrophy. Neurology. 51:1454,
1998. PMID 9818880
269. Lai Y, Yue Y, Liu M, Ghosh A, Engelhardt JF, Chamberlain JS, Duan D: Efficient in vivo gene
expression by trans-splicing adeno-associated viral vectors. Nat Biotechnol. 23:1435, 2005. PMID
16244658

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 60 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

270. Larsen UT, Juhl B, Hein-Sorensen O, de Fine Olivarius B: Complications during anaesthesia in
patients with Duchennes muscular dystrophy (a retrospective study). Can J Anaesth. 36:418, 1989.
PMID 2758541
271. Lasa A, Piccolo F, de Diego C, Jeanpierre M, Colomer J, Rodriguez MJ, Urtizberea JA, Baiget M,
Kaplan J, Gallano P: Severe limb girdle muscular dystrophy in Spanish gypsies Further evidence for a
founder mutation in the gamma-sarcoglycan gene. Eur J Hum Genet. 6:396, 1998. PMID 9781048
272. Lau KS, Grange RW, Isotani E, Sarelius IH, Kamm KE, Huang PL, Stull JT: nNOS and eNOS
modulate cGMP formation and vascular response in contracting fast-twitch skeletal muscle. Physiol
Genomics. 2:21, 2000. PMID 11015578
273. Lee JY, Qu-Petersen Z, Cao B, Kimura S, Jankowski R, Cummins J, Usas A, Gates C, Robbins P,
Wernig A, Huard J: Clonal isolation of muscle-derived cells capable of enhancing muscle regeneration
and bone healing. J Cell Biol. 150:1085, 2000.. PMID 10973997
274. Leibowitz D, Dubowitz V: Intellect and behaviour in Duchenne muscular dystrophy. Dev Med Child
Neurol. 23:577, 1981. PMID 7286451
275. Lennon NJ, Kho A, Bacskai BJ, Perlmutter SL, Hyman BT, Brown RH Jr: Dysferlin interacts with
annexins A1 and A2 and mediates sarcolemmal wound-healing. J Biol Chem. 278:50466, 2003.
PMID 14506282
276. Levine BA, Moir AJ, Patchell VB, Perry SV: Binding sites involved in the interaction of actin with the
N-terminal region of dystrophin. FEBS Lett. 298:44, 1992. PMID 1544421
277. Li J, Dressman D, Tsao YP, Sakamoto A, Hoffman EP, Xiao X: rAAV vector-mediated sarcogylcan
gene transfer in a hamster model for limb girdle muscular dystrophy. Gene Ther. 6:74, 1999. PMID
10341878
278. Li J, Wang D, Qian S, Chen Z, Zhu T, Xiao X: Efficient and long-term intracardiac gene transfer in
delta-sarcoglycan-deficiency hamster by adeno-associated virus-2 vectors. Gene Ther. 10:1807,
2003. PMID 12960970
279. Lidov HG: Dystrophin in the nervous system. Brain Pathol. 6:63, 1996. PMID 8866748
280. Lidov HG, Byers TJ, Kunkel LM: The distribution of dystrophin in the murine central nervous system
An immunocytochemical study. Neuroscience. 54:167, 1993. PMID 8515841
281. Lidov HG, Byers TJ, Watkins SC, Kunkel LM: Localization of dystrophin to postsynaptic regions of
central nervous system cortical neurons. Nature. 348:725, 1990. PMID 2259381
282. Lidov HG, Selig S, Kunkel LM: Dp140 A novel 140 kDa CNS transcript from the dystrophin locus.
Hum Mol Genet. 4:329, 1995. PMID 7795584
283. Lim LE, Duclos F, Broux O, Borg N, Sunada Y, Allamand V, Meyer J, Richard I, Moomaw C,
Slaughter C: , et al Beta-sarcoglycan. : 257, 1995. PMID 7581448
284. Little W: On the Nature and Treatment of the Deformities of the Human Frame Being a Course of
Lectures Delivered at the Royal Orthopedic Hospital in. 1843: With Numerous Notes and Additions to
the Present Time. London, Longman, Brown, Green, and Longmans, 1953. PMID 7581448
285. Liu J, Aoki M, Illa I, Wu C, Fardeau M, Angelini C, Serrano C, Urtizberea JA, Hentati F, Hamida MB,
Bohlega S, Culper EJ, Amato AA, Bossie K, Oeltjen J, Bejaoui K, McKenna-Yasek D, Hosler BA, Schurr
E, Arahata K, de Jong PJ, Brown RH Jr: Dysferlin, a novel skeletal muscle gene, is mutated in Miyoshi
myopathy and limb girdle muscular dystrophy. Nat Genet. 20:31, 1998. PMID 9731526
286. Liu J, Lissens W, van Broeckhoven C, Lofgren A, Camus M, Liebaers I, Van Steirteghem A: Normal
pregnancy after preimplantation DNA diagnosis of a dystrophin gene deletion. Prenat Diagn. 15:351,
1995. PMID 7617576
287. Liu M, Yue Y, Harper SQ, Grange RW, Chamberlain JS, Duan D: Adeno-associated virus-mediated
microdystrophin expression protects young mdx muscle from contraction-induced injury. Mol Ther.
11:245, 2005. PMID 15668136
288. Ljunggren A, Duggan D, McNally E, Boylan KB, Gama CH, Kunkel LM, Hoffman EP: Primary adhalin
deficiency as a cause of muscular dystrophy in patients with normal dystrophin. Ann Neurol. 38:367,
1995. PMID 7668821

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 61 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

289. Longman C, Brockington M, Torelli S, Jimenez-Mallebrera C, Kennedy C, Khalil N, Feng L, Saran


RK, Voit T, Merlini L, Sewry CA, Brown SC, Muntoni F: Mutations in the human LARGE gene cause
MDC1D, a novel form of congenital muscular dystrophy with severe mental retardation and abnormal
glycosylation of alpha-dystroglycan. Hum Mol Genet. 12:2853, 2003. PMID 12966029
290. Love DR, Hill DF, Dickson G, Spurr NK, Byth BC, Marsden RF, Walsh FS, Edwards YH, Davies KE:
An autosomal transcript in skeletal muscle with homology to dystrophin. Nature. 339:55, 1989. PMID
2541343
291. Lu L, Shen RN, Broxmeyer HE: Stem cells from bone marrow, umbilical cord blood and peripheral
blood for clinical application Current status and future application. Crit Rev Oncol Hematol. 22:61,
1996. PMID 8679101
292. Lu QL, Mann CJ, Lou F, Bou-Gharios G, Morris GE, Xue SA, Fletcher S, Partridge TA, Wilton SD:
Functional amounts of dystrophin produced by skipping the mutated exon in the mdx dystrophic mouse.
Nat Med. 9:1009, 2003. PMID 12847521
293. Lu QL, Rabinowitz A, Chen YC, Yokota T, Yin H, Alter J, Jadoon A, Bou-Gharios G, Partridge T:
Systemic delivery of antisense oligoribonucleotide restores dystrophin expression in body-wide skeletal
muscles. Proc Natl Acad Sci USA. 102:198, 2005. PMID 15608067
294. Lunt PW, Jardine PE, Koch MC, Maynard J, Osborn M, Williams M, Harper PS, Upadhyaya M:
Correlation between fragment size at D4F104S1 and age at onset or at wheelchair use, with a possible
generational effect, accounts for much phenotypic variation in 4q35-facioscapulohumeral muscular
dystrophy (FSHD). Hum Mol Genet. 4:951, 1995. PMID 7633457
295. Lupski JR, Garcia CA, Zoghbi HY, Hoffman EP, Fenwick RG: Discordance of muscular dystrophy in
monozygotic female twins Evidence supporting asymmetric splitting of the inner cell mass in a manifesting
carrier of Duchenne dystrophy. Am J Med Genet. 40:354, 1991. PMID 1683155
296. MacLennan DH, Duff C, Zorzato F, Fujii J, Phillips M, Korneluk RG, Frodis W, Britt BA, Worton RG:
Ryanodine receptor gene is a candidate for predisposition to malignant hyperthermia. Nature.
343:559, 1990. PMID 1967823
297. Malhotra SB, Hart KA, Klamut HJ, Thomas NS, Bodrug SE, Burghes AH, Bobrow M, Harper PS,
Thompson MW, Ray PN: , et al Frame-shift deletions in patients with Duchenne and Becker muscular
dystrophy. Science. 242:755, 1988. PMID 3055295
298. Manilal S, Nguyen TM, Morris GE: Colocalization of emerin and lamins in interphase nuclei and
changes during mitosis. Biochem Biophys Res Commun. 249:643, 1998. PMID 9731189
299. Manilal S, Nguyen TM, Sewry CA, Morris GE: The Emery-Dreifuss muscular dystrophy protein,
emerin, is a nuclear membrane protein. Hum Mol Genet. 5:801, 1996. PMID 8776595
300. Mann CJ, Honeyman K, Cheng AJ, Ly T, Lloyd F, Fletcher S, Morgan JE, Partridge TA, Wilton SD:
Antisense-induced exon skipping and synthesis of dystrophin in the mdx mouse. Proc Natl Acad Sci
USA. 98:42, 2001. PMID 11120883
301. Markesbery WR, Griggs RC, Herr B: Distal myopathy Electron microscopic and histochemical
studies. Neurology. 27:727, 1977. PMID 196233
302. Matsuda C, Hayashi YK, Ogawa M, Aoki M, Murayama K, Nishino I, Nonaka I, Arahata K, Brown RH
Jr: The sarcolemmal proteins dysferlin and caveolin-3 interact in skeletal muscle. Hum Mol Genet.
10:1761, 2001.. PMID 11532985
303. Matsumura K, Ervasti JM, Ohlendieck K, Kahl SD, Campbell KP: Association of dystrophin-related
protein with dystrophin-associated proteins in mdx mouse muscle. Nature. 360:588, 1992. PMID
1461282
304. Matthews PM, Karpati G: Pattern of X-chromosome inactivation as a key determinant of the
clinicopathologic phenotype of Duchenne muscular dystrophy carriers. Neurology. 46:1189, 1996.
PMID 8780133
305. Mayer U, Saher G, Fassler R, Bornemann A, Echtermeyer F, von der Mark H, Miosge N, Poschl E,
von der Mark K: Absence of integrin alpha 7 causes a novel form of muscular dystrophy. Nat Genet.
17:318, 1997. PMID 9354797

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 62 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

306. McArdle A, Edwards RH, Jackson MJ: Time course of changes in plasma membrane permeability in
the dystrophin-deficient mdx mouse. Muscle Nerve. 17:1378, 1994. PMID 7969238
307. McCarthy TV, Healy JM, Heffron JJ, Lehane M, Deufel T, Lehmann-Horn F, Farrall M, Johnson K:
Localization of the malignant hyperthermia susceptibility locus to human chromosome 19q12-13.2.
Nature. 343:562, 1990. PMID 2300206
308. McNally EM, Duggan D, Gorospe JR, Bonnemann CG, Fanin M, Pegoraro E, Lidov HG, Noguchi S,
Ozawa E, Finkel RS, Cruse RP, Angelini C, Kunkel LM, Hoffman EP: Mutations that disrupt the
carboxyl-terminus of gamma-sarcoglycan cause muscular dystrophy. Hum Mol Genet. 5:1841, 1996a.
PMID 8923014
309. McNally EM, Ly CT, Kunkel LM: Human epsilon-sarcoglycan is highly related to alpha-sarcoglycan
(adhalin), the limb girdle muscular dystrophy 2D gene. FEBS Lett. 422:27, 1998. PMID 9475163
310. McNally EM, Passos-Bueno MR, Bonnemann CG, Vainzof M, de Sa Moreira E, Lidov HG, Othmane
KB, Denton PH, Vance JM, Zatz M, Kunkel LM: Mild and severe muscular dystrophy caused by a single
gamma-sarcoglycan mutation. Am J Hum Genet. 59:1040, 1996b. PMID 8900232
311. McPherron AC, Lawler AM, Lee SJ: Regulation of skeletal muscle mass in mice by a new TGF-beta
superfamily member. Nature. : 83, 1997. PMID 9139826
312. McPherron AC, Lee SJ: Double muscling in cattle due to mutations in the myostatin gene. Proc Natl
Acad Sci USA. 94:12457, 1997. PMID 9356471
313. Meier H, Southard JL: Muscular dystrophy in the mouse caused by an allele at the dy-locus. Life Sci.
9:137, 1970. PMID 5434356
314. Melacini P, Fanin M, Duggan DJ, Freda MP, Berardinelli A, Danieli GA, Barchitta A, Hoffman EP,
Dalla Volta S, Angelini C: Heart involvement in muscular dystrophies due to sarcoglycan gene mutations.
Muscle Nerve. 22:473, 1999. PMID 10204782
315. Mendell JR, Kissel JT, Amato AA, King W, Signore L, Prior TW, Sahenk Z, Benson S, McAndrew PE,
Rice R: , et al Myoblast transfer in the treatment of Duchennes muscular dystrophy. N Engl J Med.
333:832, 1995. PMID 7651473
316. Mendell JR, Moxley RT, Griggs RC, Brooke MH, Fenichel GM, Miller JP, King W, Signore L, Pandya
S, Florence J: , et al Randomized, double-blind six-month trial of prednisone in Duchennes muscular
dystrophy. N Engl J Med X320X. : 1592, 1989. PMID 2657428
317. Mendell JR, Province MA, Moxley RT 3rd, Griggs RC, Brooke MH, Fenichel GM, Miller JP, Kaiser KK,
King W, Robison J: , et al Clinical investigation of Duchenne muscular dystrophy. A methodology for
therapeutic trials based on natural history controls. Arch Neurol. 44:808, 1987. PMID 3115236
318. Menke A, Jockusch H: Decreased osmotic stability of dystrophin-less muscle cells from the mdx
mouse. Nature. 349:69, 1991. PMID 1985268
319. Mercado : , et al. : FASEB J, 2006 (in press). PMID 1985268
320. Merlini L, Morandi L, Granata C, Ballestrazzi A: Bethlem myopathy Early-onset benign autosomal
dominant myopathy with contractures. Description of two new families. Neuromuscul Disord. 4:503,
1994. PMID 7881296
321. Meryon E: On granular and fatty degeneration of the voluntary muscles. Medico-Chirurgical Trans
(London). 35:73, 1852.
322. Mesa LE, Dubrovsky AL, Corderi J, Marco P, Flores D: Steroids in Duchenne muscular
dystrophy--deflazacort trial. Neuromuscul Disord. 1:261, 1991. PMID 1822804
323. Messina DN, Speer MC, Pericak-Vance MA, McNally EM: Linkage of familial dilated cardiomyopathy
with conduction defect and muscular dystrophy to chromosome 6q23. Am J Hum Genet. 61:909,
1997. PMID 9382102
324. Metzinger L, Passaquin AC, Warter JM, Poindron P: alpha-Methylprednisolone promotes skeletal
myogenesis in dystrophin-deficient and control mouse cultures. Neurosci Lett. 155:171, 1993. PMID
8377947
325. Mezon BJ, Wrogemann K, Blanchaer MC: Differing populations of mitochondria isolated from the
skeletal muscle of normal and dystrophic hamsters. Can J Biochem. 52:1024, 1974. PMID 4429865

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 63 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

326. Michele DE, Barresi R, Kanagawa M, Saito F, Cohn RD, Satz JS, Dollar J, Nishino I, Kelley RI,
Somer H, Straub V, Mathews KD, Moore SA, Campbell KP: Post-translational disruption of
dystroglycan-ligand interactions in congenital muscular dystrophies. Nature. 418:417, 2002. PMID
12140558
327. Milasin J, Muntoni F, Severini GM, Bartoloni L, Vatta M, Krajinovic M, Mateddu A, Angelini C,
Camerini F, Falaschi A, Mestroni L, Giacca M: A point mutation in the 5 splice site of the dystrophin
gene first intron responsible for X-linked dilated cardiomyopathy. Hum Mol Genet. 5:73, 1996.. PMID
8789442
328. Miller RG, Sharma KR, Pavlath GK, Gussoni E, Mynhier M, Lanctot AM, Greco CM, Steinman L, Blau
HM: Myoblast implantation in Duchenne muscular dystrophy The San Francisco study. Muscle Nerve.
20:469, 1997. PMID 9121505
329. 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. Nat Genet. 18:365, 1998.
PMID 9537420
330. Miyoshi K, Iwasa M, Kawai H, Sasaki N, Kusaka K, Yagita M, Hiasa M, Tada Y: [Autosomal recessive
distal muscular dystrophy--a new type of distal muscular dystrophy observed characteristically in Japan.
] Nippon Rinsho. 35:3922, 1977. PMID 599673
331. Miyoshi K, Kawai H, Iwasa M, Kusaka K, Nishino H: Autosomal recessive distal muscular dystrophy
as a new type of progressive muscular dystrophy. Seventeen cases in eight families including an
autopsied case. Brain. 109:31, 1986. PMID 3942856
332. Moens P, Baatsen PH, Marechal G: Increased susceptibility of EDL muscles from mdx mice to
damage induced by contractions with stretch. J Muscle Res Cell Motil. 14:446, 1993. PMID 7693747
333. Moghadaszadeh B, Petit N, Jaillard C, Brockington M, Roy SQ, Merlini L, Romero N, Estournet B,
Desguerre I, Chaigne D, Muntoni F, Topaloglu H, Guicheney P: Mutations in SEPN1 cause congenital
muscular dystrophy with spinal rigidity and restrictive respiratory syndrome. Nat Genet. 29:17, 2001.
PMID 11528383
334. Monaco AP, Bertelson CJ, Liechti-Gallati S, Moser H, Kunkel LM: An explanation for the phenotypic
differences between patients bearing partial deletions of the DMD locus. Genomics. 2:90, 1988. PMID
3384440
335. Monaco AP, Bertelson CJ, Middlesworth W, Colletti CA, Aldridge J, Fischbeck KH, Bartlett R,
Pericak-Vance MA, Roses AD, Kunkel LM: Detection of deletions spanning the Duchenne muscular
dystrophy locus using a tightly linked DNA segment. Nature. 316:842, 1985. PMID 2993910
336. Monaco AP, Neve RL, Colletti-Feener C, Bertelson CJ, Kurnit DM, Kunkel LM: Isolation of candidate
cDNAs for portions of the Duchenne muscular dystrophy gene. Nature. 323:646, 1986. PMID
3773991
337. 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. Nat Genet. 24:163, 2000. PMID 10655062
338. Moxley RT 3rd, Ashwal S, Pandya S, Connolly A, Florence J, Mathews K, Baumbach L, McDonald C,
Sussman M, Wade C: Practice parameter Corticosteroid treatment of Duchenne dystrophy. : 13,
2005. PMID 15642897
339. Mues A, van der Ven PF, Young P, Furst DO, Gautel M: Two immunoglobulin-like domains of the
Z-disc portion of titin interact in a conformation-dependent way with telethonin. FEBS Lett. 428:111,
1998. PMID 9645487
340. Muntoni F, Cau M, Ganau A, Congiu R, Arvedi G, Mateddu A, Marrosu MG, Cianchetti C, Realdi G,
Cao ABC, LASTNAME ABC, LASTNAME A: , et al Brief report. : 921, 1993. PMID 8361506
341. Muntoni F, Lichtarowicz-Krynska EJ, Sewry CA, Manilal S, Rcan D, Llense S, Taylor J, Morris GE,
Dubowitz V: Early presentation of X-linked Emery-Dreifuss muscular dystrophy resembling limb-girdle
muscular dystrophy. Neuromuscul Disord. 8:72, 1998. PMID 9608559

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 64 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

342. Murray JM, Davies KE, Harper PS, Meredith L, Mueller CR, Williamson R: Linkage relationship of a
cloned DNA sequence on the short arm of the X chromosome to Duchenne muscular dystrophy.
Nature. 300:69, 1982. PMID 6982420
343. Nagano A, Koga R, Ogawa M, Kurano Y, Kawada J, Okada R, Hayashi YK, Tsukahara T, Arahata K:
Emerin deficiency at the nuclear membrane in patients with Emery-Dreifuss muscular dystrophy. Nat
Genet. 12:254, 1996. PMID 8589715
344. Naom I, DAlessandro M, Sewry C, Ferlini A, Topaloglu H, Helbling-Leclerc A, Guicheney P, Schwartz
K, Akcoren Z, Dubowitz V, Muntoni F: The role of immunocytochemistry and linkage analysis in the
prenatal diagnosis of merosin-deficient congenital muscular dystrophy. Hum Genet. 99:535, 1997a.
PMID 9099847
345. Naom I, Sewry C, DAlessandro M, Topaloglu H, Ferlini A, Wilson L, Dubowitz V, Muntoni F: Prenatal
diagnosis in merosin-deficient congenital muscular dystrophy. Neuromuscul Disord. 7:176, 1997b.
PMID 9185181
346. Nicholson LV, Davison K, Falkous G, Harwood C, ODonnell E, Slater CR, Harris JB: Dystrophin in
skeletal muscle. I. Western blot analysis using a monoclonal antibody. J Neurol Sci. 94:125, 1989.
PMID 2693617
347. Nigro V, Bruni P, Ciccodicola A, Politano L, Nigro G, Piluso G, Cappa V, Covone AE, Romeo G,
DUrso M: SSCP detection of novel mutations in patients with Emery-Dreifuss muscular dystrophy
Definition of a small C-terminal region required for emerin function. Hum Mol Genet. 4:2003, 1995.
PMID 8595433
348. Nigro V, de Sa 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. Nat Genet. 14:195, 1996a. PMID 8841194
349. Nigro V, Okazaki Y, Belsito A, Piluso G, Matsuda Y, Politano L, Nigro G, Ventura C, Abbondanza C,
Molinari AM, Acampora D, Nishimura M, Hayashizaki Y, Puca GA: Identification of the Syrian hamster
cardiomyopathy gene. Hum Mol Genet. 6:601, 1997. PMID 9097966
350. Nigro V, Piluso G, Belsito A, Politano L, Puca AA, Papparella S, Rossi E, Viglietto G, Esposito MG,
Abbondanza C, Medici N, Molinari AM, Nigro G, Puca GA: Identification of a novel sarcoglycan gene at
5q33 encoding a sarcolemmal 35 kDa glycoprotein. Hum Mol Genet. 5:1179, 1996b. PMID 8842738
351. Nissinen M, Helbling-Leclerc A, Zhang X, Evangelista T, Topaloglu H, Cruaud C, Weissenbach J,
Fardeau M, Tome FM, Schwartz K, Tryggvason K, Guicheney P: Substitution of a conserved cysteine-996
in a cysteine-rich motif of the laminin alpha2-chain in congenital muscular dystrophy with partial deficiency
of the protein. Am J Hum Genet. 58:1177, 1996. PMID 8651294
352. Noguchi S, McNally EM, Ben Othmane K, Hagiwara Y, Mizuno Y, Yoshida M, Yamamoto H,
Bonnemann CG, Gussoni E, Denton PH: , et al Mutations in the dystrophin-associated protein
gamma-sarcoglycan in chromosome 13 muscular dystrophy. Science. 270:819, 1995. PMID 7481775
353. Nolan MA, Jones OD, Pedersen RL, Johnston HM: Cardiac assessment in childhood carriers of
Duchenne and Becker muscular dystrophies. Neuromuscul Disord. 13:129, 2003. PMID 12565910
354. Nudel U, Robzyk K, Yaffe D: Expression of the putative Duchenne muscular dystrophy gene in
differentiated myogenic cell cultures and in the brain. Nature. 331:635, 1988. PMID 3340214
355. OBrien KF, Kunkel LM: Dystrophin and muscular dystrophy Past, present, and future. Mol Genet
Metab. 74:75, 2001. PMID 11592805
356. Oak SA, Russo K, Petrucci TC, Jarrett HW: Mouse alpha1-syntrophin binding to Grb2 Further
evidence of a role for syntrophin in cell signaling. Biochemistry. 40:11270, 2001. PMID 11551227
357. Ohlendieck K, Campbell KP: Dystrophin constitutes 5% of membrane cytoskeleton in skeletal muscle.
FEBS Lett. 283:230, 1991. PMID 2044761
358. Ohlendieck K, Ervasti JM, Matsumura K, Kahl SD, Leveille CJ, Campbell KP: Dystrophin-related
protein is localized to neuromuscular junctions of adult skeletal muscle. Neuron. 7:499, 1991. PMID
1654951
359. Ohlendieck K, Matsumura K, Ionasescu VV, Towbin JA, Bosch EP, Weinstein SL, Sernett SW,

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 65 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Campbell KP: Duchenne muscular dystrophy Deficiency of dystrophin-associated proteins in the


sarcolemma. Neurology. 43:795, 1993. PMID 8469343
360. Okinaka S, Kumagai H, Ebashi S, Sugita H, Momoi H, Toyokura Y, Fujie Y: Serum creatine
phosphokinase. Activity in progressive muscular dystrophy and neuromuscular diseases. Arch
Neurol. 4:520, 1961. PMID 13730599
361. Okinaka S, Sugita H, Momoi H, Toyokura Y, Kumagai H, Ebashi S, Fujie Y: Serum creatine
phosphokinase and aldolase activity in neuromuscular disorders. Trans Am Neurol Assoc. 84:62,
1959. PMID 13854172
362. Ono Y, Shimada H, Sorimachi H, Richard I, Saido TC, Beckmann JS, Ishiura S, Suzuki K: Functional
defects of a muscle-specific calpain, p94, caused by mutations associated with limb-girdle muscular
dystrophy type 2A. J Biol Chem. 273:17073, 1998. PMID 9642272
363. Oudet C, Hanauer A, Clemens P, Caskey T, Mandel JL: Two hot spots of recombination in the DMD
gene correlate with the deletion prone regions. Hum Mol Genet. 1:599, 1992. PMID 1363782
364. Pan TC, Zhang RZ, Pericak-Vance MA, Tandan R, Fries T, Stajich JM, Viles K, Vance JM, Chu ML,
Speer MC: Missense mutation in a von Willebrand factor type A domain of the alpha 3(VI) collagen gene
(COL6A3) in a family with Bethlem myopathy. Hum Mol Genet. 7:807, 1998. PMID 9536084
365. Parsons MJ, Campos I, Hirst EM, Stemple DL: Removal of dystroglycan causes severe muscular
dystrophy in zebrafish embryos. Development. 129:3505, 2002. PMID 12091319
366. Partridge TA, Morgan JE, Coulton GR, Hoffman EP, Kunkel LM: Conversion of mdx myofibres from
dystrophin-negative to -positive by injection of normal myoblasts. Nature. 337:176, 1989. PMID
2643055
367. Passaquin AC, Metzinger L, Leger JJ, Warter JM, Poindron P: Prednisolone enhances myogenesis
and dystrophin-related protein in skeletal muscle cell cultures from mdx mouse. J Neurosci Res.
35:363, 1993. PMID 8360945
368. Pegoraro E, Mancias P, Swerdlow SH, Raikow RB, Garcia C, Marks H, Crawford T, Carver V, Di
Cianno B, Hoffman EP: Congenital muscular dystrophy with primary laminin alpha2 (merosin) deficiency
presenting as inflammatory myopathy. Ann Neurol. 40:782, 1996. PMID 8957020
369. Penisson-Besnier I, Richard I, Dubas F, Beckmann JS, Fardeau M: Pseudometabolic expression and
phenotypic variability of calpain deficiency in two siblings. Muscle Nerve. 21:1078, 1998. PMID
9655129
370. Peters MF, OBrien KF, Sadoulet-Puccio HM, Kunkel LM, Adams ME, Froehner SC:
beta-dystrobrevin, a new member of the dystrophin family. Identification, cloning, and protein associations.
J Biol Chem. 272:31561, 1997. PMID 9395493
371. Petrof BJ, Shrager JB, Stedman HH, Kelly AM, Sweeney HL: Dystrophin protects the sarcolemma
from stresses developed during muscle contraction. Proc Natl Acad Sci USA. 90:3710, 1993. PMID
8475120
372. Piccolo F, Jeanpierre M, Leturcq F, Dode C, Azibi K, Toutain A, Merlini L, Jarre L, Navarro C,
Krishnamoorthy R, Tome FM, Urtizberea JA, Beckmann JS, Campbell KP, Kaplan JC: A founder mutation
in the gamma-sarcoglycan gene of gypsies possibly predating their migration out of India. Hum Mol
Genet. 5:2019, 1996. PMID 8968757
373. Piccolo F, Roberds SL, Jeanpierre M, Leturcq F, Azibi K, Beldjord C, Carrie A, Rcan D, Chaouch
M, Reghis A: , et al Primary adhalinopathy. : 243, 1995. PMID 7663524
374. Politano L, Nigro V, Nigro G, Petretta VR, Passamano L, Papparella S, Di Somma S, Comi LI:
Development of cardiomyopathy in female carriers of Duchenne and Becker muscular dystrophies.
JAMA. 275:1335, 1996. PMID 8614119
375. Porter JD, Khanna S, Kaminski HJ, Rao JS, Merriam AP, Richmonds CR, Leahy P, Li J, Guo W,
Andrade FH: A chronic inflammatory response dominates the skeletal muscle molecular signature in
dystrophin-deficient mdx mice. Hum Mol Genet. 11:263, 2002. PMID 11823445
376. Porter JD, Merriam AP, Khanna S, Andrade FH, Richmonds CR, Leahy P, Cheng G, Karathanasis P,
Zhou X, Kusner LL, Adams ME, Willem M, Mayer U, Kaminski HJ: Constitutive properties, not molecular

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 66 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

adaptations, mediate extraocular muscle sparing in dystrophic mdx mice. FASEB J. 17:893, 2003.
PMID 12670877
377. Porter JD, Merriam AP, Leahy P, Gong B, Feuerman J, Cheng G, Khanna S: Temporal gene
expression profiling of dystrophin-deficient (mdx) mouse diaphragm identifies conserved and muscle
group-specific mechanisms in the pathogenesis of muscular dystrophy. Hum Mol Genet. 13:257,
2004. PMID 14681298
378. Prelle A, Comi GP, Tancredi L, Rigoletto C, Ciscato P, Fortunato F, Nesti S, Sciacco M, Robotti M,
Bazzi P, Felisari G, Moggio M, Scarlato G: Sarcoglycan deficiency in a large Italian population of
myopathic patients. Acta Neuropathol (Berl). 96:509, 1998. PMID 9829815
379. Puca AA, Nigro V, Piluso G, Belsito A, Sampaolo S, Quaderi N, Rossi E, Di Iorio G, Ballabio A,
Franco B: Identification and characterization of a novel member of the dystrobrevin gene family.
FEBS Lett. 425:7, 1998. PMID 9540997
380. Qu Z, Balkir L, van Deutekom JC, Robbins PD, Pruchnic R, Huard J: Development of approaches to
improve cell survival in myoblast transfer therapy. J Cell Biol. 142:1257, 1998. PMID 9732286
381. Rafael JA, Tinsley JM, Potter AC, Deconinck AE, Davies KE: Skeletal muscle-specific expression of a
utrophin transgene rescues utrophin-dystrophin deficient mice. Nat Genet. 19:79, 1998. PMID
9590295
382. 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. Am J Hum Genet. 66:1407, 2000. PMID 10739764
383. Ragot T, Stratford-Perricaudet LD, Vincent N, Chafey P, Vigne E, Gilgenkrantz H, Couton D, Briand
P, Kaplan JC, Kahn A: , et al Adenovirus-mediated transfer of a human dystrophin gene to skeletal muscle
of mdx mouse. Gene Ther 1(Suppl 1). : S53, 1994. PMID 8542399
384. Rando TA: Role of nitric oxide in the pathogenesis of muscular dystrophies A "two hit" hypothesis of
the cause of muscle necrosis. Microsc Res Tech. 55:223, 2001. PMID 11748861
385. Rapaport D, Fuchs O, Nudel U, Yaffe D: Expression of the Duchenne muscular dystrophy gene
products in embryonic stem cells and their differentiated derivatives. J Biol Chem. 267:21289, 1992.
PMID 1400438
386. Ray PN, Belfall B, Duff C, Logan C, Kean V, Thompson MW, Sylvester JE, Gorski JL, Schmickel RD,
Worton RG: Cloning of the breakpoint of an X;21 translocation associated with Duchenne muscular
dystrophy. Nature. 318:672, 1985. PMID 3001530
387. Reitter B: Deflazacort vs. prednisone in Duchenne muscular dystrophy Trends of an ongoing study.
Brain Dev 17(Suppl). : 39, 1995. PMID 8882570
388. Repetto S, Bado M, Broda P, Lucania G, Masetti E, Sotgia F, Carbone I, Pavan A, Bonilla E, Cordone
G, Lisanti MP, Minetti C: Increased number of caveolae and caveolin-3 overexpression in Duchenne
muscular dystrophy. Biochem Biophys Res Commun. 261:547, 1999. PMID 10441463
389. Richard I, Brenguier L, Dincer P, Rodat C, Bady B, Burgunder JM, Chemaly R, Garcia CA,
Halaby G, Jackson CE, Kurnit DM, Lefranc G, Legum C, Loiselet J, Merlini L, Nivelon-Chevallier A,
Ollagnon-Roman E, Restagno G, Topaloglu H, Beckmann JS: Multiple independent molecular etiology for
limb-girdle muscular dystrophy type 2A patients from various geographical origins. Am J Hum Genet.
60:1128, 1997. PMID 9150160
390. Richard I, Broux O, Allamand V, Fougerousse F, Chiannilkulchai N, Borg N, Brenguier L, Devaud
C, Pasturaud P, Rodat C: , et al Mutations in the proteolytic enzyme calpain 3 cause limb-girdle
muscular dystrophy type 2A. Cell. 81:27, 1995. PMID 7720071
391. Richards CS, Watkins SC, Hoffman EP, Schneider NR, Milsark IW, Katz KS, Cook JD, Kunkel LM,
Cortada JM: Skewed X inactivation in a female MZ twin results in Duchenne muscular dystrophy. Am
J Hum Genet. 46:672, 1990. PMID 2180286
392. Rifai Z, Welle S, Moxley RT 3rd, Lorenson M, Griggs RC: Effect of prednisone on protein metabolism
in Duchenne dystrophy. Am J Physiol. 268:E67, 1995. PMID 7840185

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 67 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

393. Roberds SL, Anderson RD, Ibraghimov-Beskrovnaya O, Campbell KP: Primary structure and
muscle-specific expression of the 50-kDa dystrophin-associated glycoprotein (adhalin). J Biol Chem.
268:23739, 1993a. PMID 8226900
394. Roberds SL, Ervasti JM, Anderson RD, Ohlendieck K, Kahl SD, Zoloto D, Campbell KP: Disruption of
the dystrophin-glycoprotein complex in the cardiomyopathic hamster. J Biol Chem. 268:11496,
1993b. PMID 8505286
395. Roberds SL, Leturcq F, Allamand V, Piccolo F, Jeanpierre M, Anderson RD, Lim LE, Lee JC, Tome
FM, Romero NB: , et al Missense mutations in the adhalin gene linked to autosomal recessive muscular
dystrophy. Cell. 78:625, 1994. PMID 8069911
396. Roberts RG, Coffey AJ, Bobrow M, Bentley DR: Exon structure of the human dystrophin gene.
Genomics. 16:536, 1993. PMID 8314593
397. Roberts RG, Gardner RJ, Bobrow M: Searching for the 1 in 2,400,000 A review of dystrophin gene
point mutations. Hum Mutat. 4:1, 1994. PMID 7951253
398. Roques I, Pedespan JM, Boisserie-Lacroix V, Ferrer X, Fontan D: [Facioscapulohumeral myopathy
and germinal mosaicism.]. Arch Pediatr. 5:880, 1998. PMID 9759294
399. Rouger K, Le Cunff M, Steenman M, Potier MC, Gibelin N, Dechesne CA, Leger JJ: Global/temporal
gene expression in diaphragm and hindlimb muscles of dystrophin-deficient (mdx) mice. Am J Physiol
Cell Physiol. 283:C773, 2002. PMID 12176734
400. Rybakova IN, Amann KJ, Ervasti JM: A new model for the interaction of dystrophin with F-actin. J Cell
Biol. 135:661, 1996. PMID 8909541
401. Rybakova IN, Ervasti JM: Dystrophin-glycoprotein complex is monomeric and stabilizes actin
filaments in vitro through a lateral association. J Biol Chem. 272:28771, 1997. PMID 9353348
402. Ryder-Cook AS, Sicinski P, Thomas K, Davies KE, Worton RG, Barnard EA, Darlison MG, Barnard
PJ: Localization of the mdx mutation within the mouse dystrophin gene. Embo J. 7:3017, 1988. PMID
2903046
403. Sadoulet-Puccio HM, Khurana TS, Cohen JB, Kunkel LM: Cloning and characterization of the human
homologue of a dystrophin related phosphoprotein found at the Torpedo electric organ post-synaptic
membrane. Hum Mol Genet. 5:489, 1996. PMID 8845841
404. Sadoulet-Puccio HM, Rajala M, Kunkel LM: Dystrobrevin and dystrophin An interaction through
coiled-coil motifs. Proc Natl Acad Sci USA. 94:12413, 1997. PMID 9356463
405. Saito K, Osawa M, Wang ZP, Ikeya K, Fukuyama Y, Kondo-Iida E, Toda T, Ohashi H, Kurosawa K,
Wakai S, Kaneko K: Haplotype-phenotype correlation in Fukuyama congenital muscular dystrophy.
Am J Med Genet. 92:184, 2000. PMID 10817652
406. Sakamoto A, Ono K, Abe M, Jasmin G, Eki T, Murakami Y, Masaki T, Toyo-Oka T, Hanaoka F: Both
hypertrophic and dilated cardiomyopathies are caused by mutation of the same gene, delta-sarcoglycan,
in hamster An animal model of disrupted dystrophin-associated glycoprotein complex. Proc Natl Acad
Sci USA. 94:13873, 1997. PMID 9391120
407. Salmikangas P, Mykkanen OM, Gronholm M, Heiska L, Kere J, Carpn O: Myotilin, a novel
sarcomeric protein with two Ig-like domains, is encoded by a candidate gene for limb-girdle muscular
dystrophy. Hum Mol Genet. 8:1329, 1999. PMID 10369880
408. Salmikangas P, van der Ven PF, Lalowski M, Taivainen A, Zhao F, Suila H, Schroder R, Lappalainen
P, Furst DO, Carpn O: Myotilin, the limb-girdle muscular dystrophy 1A (LGMD1A) protein, cross-links
actin filaments and controls sarcomere assembly. Hum Mol Genet. 12:189, 2003. PMID 12499399
409. Sampaolesi M, Torrente Y, Innocenzi A, Tonlorenzi R, DAntona G, Pellegrino MA, Barresi R,
Bresolin N, De Angelis MG, Campbell KP, Bottinelli R, Cossu G: Cell therapy of alpha-sarcoglycan null
dystrophic mice through intra-arterial delivery of mesoangioblasts. Science. 301:487, 2003. PMID
12855815
410. Schuelke M, Wagner KR, Stolz LE, Hubner C, Riebel T, Komen W, Braun T, Tobin JF, Lee SJ:
Myostatin mutation associated with gross muscle hypertrophy in a child. N Engl J Med. 350:2682,
2004. PMID 15215484

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 68 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

411. Schwartz M, Hertz JM, Sveen ML, Vissing J: LGMD2I presenting with a characteristic Duchenne or
Becker muscular dystrophy phenotype. Neurology. 64:1635, 2005. PMID 15883334
412. Seale P, Rudnicki MA: A new look at the origin, function, and "stem-cell" status of muscle satellite
cells. Dev Biol. 218:115, 2000. PMID 10656756
413. Sekizawa A, Kimura T, Sasaki M, Nakamura S, Kobayashi R, Sato T: Prenatal diagnosis of
Duchenne muscular dystrophy using a single fetal nucleated erythrocyte in maternal blood.
Neurology. 46:1350, 1996. PMID 8628481
414. Sewry CA, Naom I, DAlessandro M, Sorokin L, Bruno S, Wilson LA, Dubowitz V, Muntoni F: Variable
clinical phenotype in merosin-deficient congenital muscular dystrophy associated with differential
immunolabelling of two fragments of the laminin alpha 2 chain. Neuromuscul Disord. 7:169, 1997.
PMID 9185180
415. Sharp NJ, Kornegay JN, Van Camp SD, Herbstreith MH, Secore SL, Kettle S, Hung WY,
Constantinou CD, Dykstra MJ, Roses AD: , et al An error in dystrophin mRNA processing in golden
retriever muscular dystrophy, an animal homologue of Duchenne muscular dystrophy. Genomics.
13:115, 1992. PMID 1577476
416. Sibley JA, Lehninger AL: Aldolase in the serum and tissues of tumor-bearing animals. J Natl Cancer
Inst 9 303, 1949
417. Sicinski P, Geng Y, Ryder-Cook AS, Barnard EA, Darlison MG, Barnard PJ: The molecular basis of
muscular dystrophy in the mdx mouse A point mutation. Science. 244:1578, 1989. PMID 2662404
418. Simonds AK, Muntoni F, Heather S, Fielding S: Impact of nasal ventilation on survival in hypercapnic
Duchenne muscular dystrophy. Thorax. 53:949, 1998. PMID 10193393
419. Sklar RM, Brown RH Jr: Methylprednisolone increases dystrophin levels by inhibiting myotube death
during myogenesis of normal human muscle in vitro. J Neurol Sci. 101:73, 1991. PMID 2027030
420. Small K, Iber J, Warren ST: Emerin deletion reveals a common X-chromosome inversion mediated by
inverted repeats. Nat Genet. 16:96, 1997. PMID 9140403
421. Small K, Warren ST: Emerin deletions occurring on both Xq28 inversion backgrounds. Hum Mol
Genet. 7:135, 1998. PMID 9384614
422. Soltan HC, Blanchaer MC: Activity of serum aldolase and lactic dehydrogenase in patients affected
with Duchenne muscular dystrophy and in their immediate relatives. J Pediatr. 54:27, 1959. PMID
13611611
423. Song KS, Scherer PE, Tang Z, Okamoto T, Li S, Chafel M, Chu C, Kohtz DS, Lisanti MP: Expression
of caveolin-3 in skeletal, cardiac, and smooth muscle cells. Caveolin-3 is a component of the sarcolemma
and co-fractionates with dystrophin and dystrophin-associated glycoproteins. J Biol Chem.
271:15160, 1996. PMID 8663016
424. Sotgia F, Lee JK, Das K, Bedford M, Petrucci TC, Macioce P, Sargiacomo M, Bricarelli FD, Minetti C,
Sudol M, Lisanti MP: Caveolin-3 directly interacts with the C-terminal tail of beta -dystroglycan.
Identification of a central WW-like domain within caveolin family members. J Biol Chem. 275:38048,
2000. PMID 10988290
425. Speer MC, Vance JM, Grubber JM, Lennon Graham F, Stajich JM, Viles KD, Rogala A, McMichael R,
Chutkow J, Goldsmith C, Tim RW, Pericak-Vance MA: Identification of a new autosomal dominant
limb-girdle muscular dystrophy locus on chromosome 7. Am J Hum Genet. 64:556, 1999. PMID
9973293
426. Spencer MJ, Tidball JG, Anderson LV, Bushby KM, Harris JB, Passos-Bueno MR, Somer H, Vainzof
M, Zatz M: Absence of calpain 3 in a form of limb-girdle muscular dystrophy (LGMD2A). J Neurol Sci.
146:173, 1997. PMID 9077514
427. Stedman HH, Sweeney HL, Shrager JB, Maguire HC, Panettieri RA, Petrof B, Narusawa M,
Leferovich JM, Sladky JT, Kelly AM: The mdx mouse diaphragm reproduces the degenerative changes of
Duchenne muscular dystrophy. Nature. 352:536, 1991. PMID 1865908
428. Straub V, Ettinger AJ, Durbeej M, Venzke DP, Cutshall S, Sanes JR, Campbell KP:
epsilon-sarcoglycan replaces alpha-sarcoglycan in smooth muscle to form a unique

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 69 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

dystrophin-glycoprotein complex. J Biol Chem. 274:27989, 1999. PMID 10488149


429. Sunada Y, Bernier SM, Kozak CA, Yamada Y, Campbell KP: Deficiency of merosin in dystrophic dy
mice and genetic linkage of laminin M chain gene to dy locus. J Biol Chem. 269:13729, 1994. PMID
8188645
430. Sunada Y, Bernier SM, Utani A, Yamada Y, Campbell KP: Identification of a novel mutant transcript
of laminin alpha 2 chain gene responsible for muscular dystrophy and dysmyelination in dy2J mice.
Hum Mol Genet. 4:1055, 1995. PMID 7655459
431. Suzuki A, Yoshida M, Ozawa E: Mammalian alpha 1- and beta 1-syntrophin bind to the alternative
splice-prone region of the dystrophin COOH terminus. J Cell Biol. 128:373, 1995. PMID 7844151
432. Takano A, Bonnemann CG, Honda H, Sakai M, Feener CA, Kunkel LM, Sobue G: Intrafamilial
phenotypic variation in limb-girdle muscular dystrophy type 2C with compound heterozygous mutations.
Muscle Nerve. 23:807, 2000. PMID 10797406
433. Tang Z, Scherer PE, Okamoto T, Song K, Chu C, Kohtz DS, Nishimoto I, Lodish HF, Lisanti MP:
Molecular cloning of caveolin-3, a novel member of the caveolin gene family expressed predominantly in
muscle. J Biol Chem. 271:2255, 1996. PMID 8567687
434. Tawil R, McDermott MP, Pandya S, King W, Kissel J, Mendell JR, Griggs RC: A pilot trial of
prednisone in facioscapulohumeral muscular dystrophy. FSH-DY Group. Neurology. 48:46, 1997.
PMID 9008492
435. Taylor EW: Progressive vagus-glossopharyngeal paralysis with ptosis A contribution to the group of
family diseases. J Nerv Ment Dis. 42:129, 1915.
436. Tennyson CN, Klamut HJ, Worton RG: The human dystrophin gene requires 16 hours to be
transcribed and is cotranscriptionally spliced. Nat Genet. 9:184, 1995. PMID 7719347
437. Thompson TG, Chan YM, Hack AA, Brosius M, Rajala M, Lidov HG, McNally EM, Watkins S, Kunkel
LM: Filamin 2 (FLN2) A muscle-specific sarcoglycan interacting protein. J Cell Biol. 148:115, 2000.
PMID 10629222
438. Tkatchenko AV, Le Cam G, Leger JJ, Dechesne CA: Large-scale analysis of differential gene
expression in the hindlimb muscles and diaphragm of mdx mouse. Biochim Biophys Acta. 1500:17,
2000. PMID 10564714
439. Tome FM, Evangelista T, Leclerc A, Sunada Y, Manole E, Estournet B, Barois A, Campbell KP,
Fardeau M: Congenital muscular dystrophy with merosin deficiency. C R Acad Sci III. 317:351, 1994.
PMID 8000914
440. Tome FM, Fardeau M: Nuclear inclusions in oculopharyngeal dystrophy. Acta Neuropathol (Berl).
49:85, 1980. PMID 6243839
441. Topaloglu H, Dincer P, Richard I, Akcoren Z, Alehan D, Ozme S, Caglar M, Karaduman A, Urtizberea
JA, Beckmann JS: Calpain-3 deficiency causes a mild muscular dystrophy in childhood.
Neuropediatrics. 28:212, 1997. PMID 9309711
442. Tremblay JP, Bouchard JP, Malouin F, Theau D, Cottrell F, Collin H, Rouche A, Gilgenkrantz S,
Abbadi N, Tremblay M: , et al Myoblast transplantation between monozygotic twin girl carriers of
Duchenne muscular dystrophy. Neuromuscul Disord. 3:583, 1993. PMID 8186717
443. Tseng BS, Zhao P, Pattison JS, Gordon SE, Granchelli JA, Madsen RW, Folk LC, Hoffman EP, Booth
FW: Regenerated mdx mouse skeletal muscle shows differential mRNA expression. J Appl Physiol.
93:537, 2002. PMID 12133862
444. Tupler R, Barbierato L, Memmi M, Sewry CA, De Grandis D, Maraschio P, Tiepolo L, Ferlini A:
Identical de novo mutation at the D4F104S1 locus in monozygotic male twins affected by
facioscapulohumeral muscular dystrophy (FSHD) with different clinical expression. J Med Genet.
35:778, 1998. PMID 9733041
445. Tyler KL: Origins and early descriptions of "Duchenne muscular dystrophy". Muscle Nerve. 28:402,
2003. PMID 14506712
446. Udd B, Partanen J, Halonen P, Falck B, Hakamies L, Heikkila H, Ingo S, Kalimo H, Kriinen H,
Laulumaa V: , et al Tibial muscular dystrophy. Late adult-onset distal myopathy in 66 Finnish patients.

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 70 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

Arch Neurol. 50:604, 1993. PMID 8503797


447. Upadhyaya M, Lunt P, Sarfarazi M, Broadhead W, Farnham J, Harper PS: The mapping of
chromosome 4q markers in relation to facioscapulohumeral muscular dystrophy (FSHD). Am J Hum
Genet. 51:404, 1992. PMID 1642238
448. Upadhyaya M, Maynard J, Rogers MT, Lunt PW, Jardine P, Ravine D, Harper PS: Improved
molecular diagnosis of facioscapulohumeral muscular dystrophy (FSHD) Validation of the differential
double digestion for FSHD. J Med Genet. 34:476, 1997. PMID 9192267
449. Vainzof M, Passos-Bueno MR, Canovas M, Moreira ES, Pavanello RC, Marie SK, Anderson LV,
Bonnemann CG, McNally EM, Nigro V, Kunkel LM, Zatz M: The sarcoglycan complex in the six autosomal
recessive limb-girdle muscular dystrophies. Hum Mol Genet. 5:1963, 1996. PMID 8968750
450. Valentine BA, Cooper BJ, Cummings JF, de Lahunta A: Canine X-linked muscular dystrophy
Morphologic lesions. J Neurol Sci. 97:1, 1990. PMID 2370557
451. van der Ven PF, Wiesner S, Salmikangas P, Auerbach D, Himmel M, Kempa S, Hayess K, Pacholsky
D, Taivainen A, Schroder R, Carpn O, Furst DO: Indications for a novel muscular dystrophy pathway.
gamma-filamin, the muscle-specific filamin isoform, interacts with myotilin. J Cell Biol. 151:235, 2000.
PMID 11038172
452. van Deutekom JC, Wijmenga C, van Tienhoven EA, Gruter AM, Hewitt JE, Padberg GW, van Ommen
GJ, Hofker MH, Frants RR: FSHD associated DNA rearrangements are due to deletions of integral copies
of a 3.2 kb tandemly repeated unit. Hum Mol Genet. 2:2037, 1993. PMID 8111371
453. van Essen AJ, Abbs S, Baiget M, Bakker E, Boileau C, van Broeckhoven C, Bushby K, Clarke A,
Claustres M, Covone AE: , et al Parental origin and germline mosaicism of deletions and duplications
of the dystrophin gene. : 249, 1992. PMID 1733826
454. Verellen-Dumoulin C, Freund M, De Meyer R, Laterre C, Frederic J, Thompson MW, Markovic VD,
Worton RG: Expression of an X-linked muscular dystrophy in a female due to translocation involving Xp21
and non-random inactivation of the normal X chromosome. Hum Genet. 67:115, 1984. PMID
6745920
455. Victor M, Hayes R, Adams RD: Oculopharyngeal muscular dystrophy. A familial disease of late life
characterized by dysphagia and progressive ptosis of the evelids. N Engl J Med. 267:1267, 1962.
PMID 13997067
456. Villanova M, Malandrini A, Toti P, Salvestroni R, Six J, Martin JJ, Guazzi GC: Localization of merosin
in the normal human brain Implications for congenital muscular dystrophy with merosin deficiency. J
Submicrosc Cytol Pathol. 28:1, 1996. PMID 8929621
457. Vockley J, Rinaldo P, Bennett MJ, Matern D, Vladutiu GD: Synergistic heterozygosity Disease
resulting from multiple partial defects in one or more metabolic pathways. Mol Genet Metab. 71:10,
2000.. PMID 11001791
458. Voit T, Fardeau M, Tome FM: Prenatal detection of merosin expression in human placenta.
Neuropediatrics. 25:332, 1994.. PMID 7770133
459. Voit T, Haas K, Leger JO, Pons F, Leger JJ: Xp21 dystrophin and 6q dystrophin-related protein.
Comparative immunolocalization using multiple antibodies. Am J Pathol. 139:969, 1991. PMID
1951635
460. Wahle E: A novel poly(A)-binding protein acts as a specificity factor in the second phase of
messenger RNA polyadenylation. Cell. 66:759, 1991. PMID 1878970
461. Wang B, Li J, Xiao X: Adeno-associated virus vector carrying human minidystrophin genes effectively
ameliorates muscular dystrophy in mdx mouse model. Proc Natl Acad Sci USA. 97:13714, 2000.
PMID 11095710
462. Watkins SC, Hoffman EP, Slayter HS, Kunkel LM: Immunoelectron microscopic localization of
dystrophin in myofibres. Nature. 333:863, 1988. PMID 3290684
463. Weiler T, Bashir R, Anderson LV, Davison K, Moss JA, Britton S, Nylen E, Keers S, Vafiadaki E,
Greenberg CR, Bushby CR, Wrogemann K: Identical mutation in patients with limb girdle muscular
dystrophy type 2B or Miyoshi myopathy suggests a role for modifier gene(s). Hum Mol Genet. 8:871,

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 71 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

1999. PMID 10196377


464. Weller B, Karpati G, Carpenter S: Dystrophin-deficient mdx muscle fibers are preferentially vulnerable
to necrosis induced by experimental lengthening contractions. J Neurol Sci. 100:9, 1990. PMID
2089145
465. Wells DJ, Wells KE, Walsh FS, Davies KE, Goldspink G, Love DR, Chan-Thomas P, Dunckley MG,
Piper T, Dickson G: Human dystrophin expression corrects the myopathic phenotype in transgenic mdx
mice. Hum Mol Genet. 1:35, 1992. PMID 1301134
466. Whittemore LA, Song K, Li X, Aghajanian J, Davies M, Girgenrath S, Hill JJ, Jalenak M, Kelley P,
Knight A, Maylor R, OHara D, Pearson A, Quazi A, Ryerson S, Tan XY, Tomkinson KN, Veldman GM,
Widom A, Wright JF, Wudyka S, Zhao L, Wolfman NM: Inhibition of myostatin in adult mice increases
skeletal muscle mass and strength. Biochem Biophys Res Commun. 300:965, 2003. PMID 12559968
467. Wiberg C, Hedbom E, Khairullina A, Lamande SR, Oldberg A, Timpl R, Morgelin M, Heinegard D:
Biglycan and decorin bind close to the n-terminal region of the collagen VI triple helix. J Biol Chem.
276:18947, 2001. PMID 11259413
468. Wiberg C, Heinegard D, Wenglen C, Timpl R, Morgelin M: Biglycan organizes collagen VI into
hexagonal-like networks resembling tissue structures. J Biol Chem. 277:49120, 2002. PMID
12354766
469. Wijmenga C, Frants RR, Brouwer OF, Moerer P, Weber JL, Padberg GW: Location of
facioscapulohumeral muscular dystrophy gene on chromosome 4. Lancet. 336:651, 1990. PMID
1975852
470. Wijmenga C, Padberg GW, Moerer P, Wiegant J, Liem L, Brouwer OF, Milner EC, Weber JL, van
Ommen GB, Sandkuyl LA: , et al Mapping of facioscapulohumeral muscular dystrophy gene to
chromosome 4q35-qter by multipoint linkage analysis and in situ hybridization. Genomics. 9:570,
1991. PMID 2037288
471. Williamson RA, Henry MD, Daniels KJ, Hrstka RF, Lee JC, Sunada Y, Ibraghimov-Beskrovnaya O,
Campbell KP: Dystroglycan is essential for early embryonic development Disruption of Reicherts
membrane in Dag1-null mice. Hum Mol Genet. 6:831, 1997. PMID 9175728
472. Worton RG, Duff C, Sylvester JE, Schmickel RD, Willard HF: Duchenne muscular dystrophy involving
translocation of the dmd gene next to ribosomal RNA genes. Science. 224:1447, 1984. PMID
6729462
473. Wrogemann K, Blanchaer MC, Jacobson BE: A calcium-associated magnesium-responsive defect of
respiration and oxidative phosphorylation by skeletal muscle mitochondria of BIO 14.6 dystrophic
hamsters. Life Sci II. 9:1167, 1970. PMID 5477798
474. Wrogemann K, Jacobson BE, Blanchaer MC: On the mechanism of a calcium-associated defect of
oxidative phosphorylation in progessive muscular dystrophy. Arch Biochem Biophys. 159:267, 1973.
PMID 4361547
475. Wu JY, Kuban KC, Allred E, Shapiro F, Darras BT: Association of Duchenne muscular dystrophy with
autism spectrum disorder. J Child Neurol. 20:790, 2005. PMID 16417872
476. Wulff K, Ebener U, Wehnert CS, Ward PA, Reuner U, Hiebsch W, Herrmann FH, Wehnert M: Direct
molecular genetic diagnosis and heterozygote identification in X-linked Emery-Dreifuss muscular
dystrophy by heteroduplex analysis. Dis Markers. 13:77, 1997a. PMID 9160182
477. Wulff K, Parrish JE, Herrmann FH, Wehnert M: Six novel mutations in the emerin gene causing
X-linked Emery-Dreifuss muscular dystrophy. Hum Mutat. 9:526, 1997b. PMID 9195226
478. Xiao X, Li J, Samulski RJ: Efficient long-term gene transfer into muscle tissue of immunocompetent
mice by adeno-associated virus vector. J Virol. 70:8098, 1996. PMID 8892935
479. Xiao X, Li J, Tsao YP, Dressman D, Hoffman EP, Watchko JF: Full functional rescue of a complete
muscle (TA) in dystrophic hamsters by adeno-associated virus vector-directed gene therapy. J Virol.
74:1436, 2000. PMID 10627554
480. Xu H, Christmas P, Wu XR, Wewer UM, Engvall E: Defective muscle basement membrane and lack
of M-laminin in the dystrophic dy/dy mouse. Proc Natl Acad Sci USA. 91:572, 1994a. PMID 8202529

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 72 -

Any use is subject to the Terms of Use on www.ommbid.com.

Chapter 216: The Muscular Dystrophies

481. Xu H, Wu XR, Wewer UM, Engvall E: Murine muscular dystrophy caused by a mutation in the laminin
alpha 2 (Lama2) gene. Nat Genet. 8:297, 1994b. PMID 7874173
482. Yaffe D, Makover A, Lederfein D, Rapaport D, Bar S, Barnea E, Nudel U: Multiple products of the
Duchenne muscular dystrophy gene. Symp Soc Exp Biol. 46:179, 1992. PMID 1341034
483. Yamada T, Kobayashi T: A novel emerin mutation in a Japanese patient with Emery-Dreifuss
muscular dystrophy. Hum Genet. 97:693, 1996. PMID 8655156
484. Yang B, Jung D, Motto D, Meyer J, Koretzky G, Campbell KP: SH3 domain-mediated interaction of
dystroglycan and Grb2. J Biol Chem. 270:11711, 1995. PMID 7744812
485. Yoshida A, Kobayashi K, Manya H, Taniguchi K, Kano H, Mizuno M, Inazu T, Mitsuhashi H,
Takahashi S, Takeuchi M, Herrmann R, Straub V, Talim B, Voit T, Topaloglu H, Toda T, Endo T: Muscular
dystrophy and neuronal migration disorder caused by mutations in a glycosyltransferase, POMGnT1.
Dev Cell. 1:. PMID 11709191
486. Yoshida M, Ozawa E: Glycoprotein complex anchoring dystrophin to sarcolemma. J Biochem
(Tokyo). 108:748, 1990. PMID 2081733
487. Yoshimura M, Sakamoto M, Ikemoto M, Mochizuki Y, Yuasa K, Miyagoe-Suzuki Y, Takeda S: AAV
vector-mediated microdystrophin expression in a relatively small percentage of mdx myofibers improved
the mdx phenotype. Mol Ther. 10:821, 2004. PMID 15509500
488. Yoshioka M, Kuroki S: Clinical spectrum and genetic studies of Fukuyama congenital muscular
dystrophy. Am J Med Genet. 53:245, 1994. PMID 7856660
489. Yoshioka M, Toda T, Kuroki S, Hamano K: Broader clinical spectrum of Fukuyama-type congenital
muscular dystrophy manifested by haplotype analysis. J Child Neurol. 14:711, 1999. PMID 10593547
490. Yue Y, Li Z, Harper SQ, Davisson RL, Chamberlain JS, Duan D: Microdystrophin gene therapy of
cardiomyopathy restores dystrophin-glycoprotein complex and improves sarcolemma integrity in the mdx
mouse heart. Circulation. 108:1626, 2003. PMID 12952841
491. Zatz M, Marie SK, Cerqueira A, Vainzof M, Pavanello RC, Passos-Bueno MR: The
facioscapulohumeral muscular dystrophy (FSHD1) gene affects males more severely and more frequently
than females. Am J Med Genet X77X. : 155, 1998.. PMID 9605290
492. Zhu T, Zhou L, Mori S, Wang Z, McTiernan CF, Qiao C, Chen C, Wang DW, Li J, Xiao X: Sustained
whole-body functional rescue in congestive heart failure and muscular dystrophy hamsters by systemic
gene transfer. Circulation. 112:2650, 2005. PMID 16230483
493. Zhu X, Hadhazy M, Wehling M, Tidball JG, McNally EM: Dominant negative myostatin produces
hypertrophy without hyperplasia in muscle. FEBS Lett. 474:71, 2000.. PMID 10828454
494. Zimprich A, Grabowski M, Asmus F, Naumann M, Berg D, Bertram M, Scheidtmann K, Kern P,
Winkelmann J, Muller-Myhsok B, Riedel L, Bauer M, Mller T, Castro M, Meitinger T, Strom TM, Gasser
T: Mutations in the gene encoding epsilon-sarcoglycan cause myoclonus-dystonia syndrome. Nat
Genet. 29:66, 2001. PMID 11528394
495. Zubrzycka-Gaarn EE, Bulman DE, Karpati G, Burghes AH, Belfall B, Klamut HJ, Talbot J, hodges RS,
Ray PN, Worton RG: The Duchenne muscular dystrophy gene product is localized in sarcolemma of
human skeletal muscle. Nature. 333:466, 1988. PMID 3287171

Copyright The McGraw-Hill Companies, Inc. All rights reserved.

- 73 -

Any use is subject to the Terms of Use on www.ommbid.com.

Das könnte Ihnen auch gefallen