Lamperti 2010

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

978 Current Pharmaceutical Design, 2010, 16, 978-987

Muscular Dystrophies: Histology, Immunohistochemistry, Molecular


Genetics and Management

Costanza Lamperti#,* and Maurizio Moggio°

#
UO Neurogenetica Molecolare, Fondazione IRCCS Isitituto Neurologico C. Besta, Via Celoria 11, 20100 Milano Italy, °UOS
Diagnostica Neuromuscoalre Fondazione Ospedale Maggiore Policlinico, Mangiagalli Regina Elena, IRCCS via F Sforza 34, 20122
Milano, Italy

Abstract: Muscle degeneration and regeneration are two of the most evident pathological events characterizing muscular diseases and in
particular muscular dystrophies. Muscular dystrophies are an heterogeneous group of hereditary diseases affecting both children and
adults, and are characterized by muscle wasting and weakness. Until now at least 30 different genes have been associated with muscular
dystrophies. They have been divided into several subgroups depending on the distribution of the muscle weakness. Thus, the
histopathological markers of all these forms are dystrophic changes at the muscle biopsy characterized by fiber size variability, fibres
necrosis, regeneration, inflammation and connective tissues deposition. As for now, no effective therapy is available for these diseases
but new inside has now been expanded in regenerative therapy such as cell therapy and gene therapy. This review is focused on muscular
dystrophies and new acknowledgments in regenerative therapy.
Keywords: Dystrophies, theraphy, exon skipping, LGMD, CMD.

INTRODUCTION present along with replacement of muscle by connective tissue and


Muscle degeneration and regeneration are two of the most fat. The major advances over the last two decades due to the
important pathomechanisms in muscle diseases and in particular identification of numerous sarcolemmal and extracellular matrix
they are involved in the pathogenesis of muscular dystrophies. (Fig. 1) proteins and associated genes, have improved diagnostic
Since now mutations in at least 30 different genes have been precision by standardizing both the immunhistochemestry and the
associated with these diseases involving different proteins in the genetic tests, in the view of genetic therapy. As for now, no therapy
muscle membrane and in the extracellular matrix. Whichever is the has been identified but the potential role of promoting muscle
protein involved, the “via ultima” is the dystrophic aspect of muscle degeneration and restoring correct genes expression have been
tissues, that is characterized by the degeneration of the muscle studied. This review article is focused on muscular dystrophy
fibres and a push to regeneration, inflammation and the substi- empathizing the diagnosis and new prospective in treatment.
tutions of contractile tissue with connective tissue that causes the
MUSCULAR DYSTROPHIES
loss of muscle strength and muscle mass in patients. All these pro-
teins are part of the muscle membrane, such as dystrophyn, Muscular dystrophies are classified as Congenital Muscular
sarcolgycans , caveolin, integrins and dysferlin, or part of the Dystrophies (CMD), dystrophinopathies, including Duchenne
extracellular matrix such as alpha dystrolgycan, merosins, collagen Muscular Dystrophy (DMD) and Becker Muscular Dystrophy
VI or the nuclear envelop such as lamins and emerins. Never- (BMD), and Limb Girdle Muscular Dystrophies (LGMD).
theless, although a large number of genes have been identified, the
mechanism leading to the degeneration and the regeneration of the Congenital Muscular Dystrophies
muscle fibres and the subsequent deposition of connective tissues, Congenital Muscular Dystrophies (CMD) are an heterogeneous
is still not known. Muscular dystrophies are an heterogeneous group of autosomal recessive disorders [1] presenting in infancy
group of muscle diseases that are characterized by muscle weak- and characterized by muscle weakness, contractures and dystrophic
ness, and depend on an alteration of specific muscle membrane or changes at the skeletal muscle biopsy (Table 1). The association of
extracellular matrix protein. Clinically, they are characterized by mental retardation with central nervous system (CNS) abnor-
progressive muscle wasting and weakness, with a wide clinical malities, such as type II lyssencephaly, brainstem and cerebellar
heterogeneity: some of them have perinatal onset, while others alterations and eye abnormalities, is often present in Muscle Eye
affect only adults. They could progress rapidly or be associated Brain disease (MEB), Walker Warburg syndrome (WWS),
with prolonged periods of stability; finally they could be associated Fukuyama Congenital Muscular Dystrophy, or LAMA 2 deficiency.
with multisystem involvement including cardiac and central These brain alterations can be secondary to an alteration of alpha
nervous systems. Usually patients become dependent to wheelchair dystroglycan glycosylation during brain development. Recently, an
and death is caused by respiratory failure secondary to respiratory alteration of alpha dystroglycan glycosylation in brain has been
and diaphragm muscle weakness. The biochemical tests show an described in one patients with a CMD [2]. In some cases, as well as
increase in the level of creatinkinase, even 10 times the normal in the Ullrich syndrome or in the less severe Bethlem syndrome, a
value, and the EMG usually shows myopathic change. The muscle distal joint laxity can be the peculiar feature [3]. Instead, a milder
biopsy is essential to the diagnosis. As said before, all these phenotype showing only muscle weakness is characteristic for the
conditions share common histological features of ‘‘dystrophic’’ CMD1C [1].
muscle biopsy changes, including fibre size variability, muscle fibre Creatine kinase (CK) could be markedly elevated or quite
degeneration and regeneration. Inflammatory changes may be normal. Nerve conduction studies may show demyelinating neuro-
pathy in merosin-deficient CMD [4] while electromyography
*Address correspondence to this author at the UO Neurogenetica (EMG) is myopathic in quite all these forms [5] in which respira-
Molecolare, Fondazione IRCCS Isitituto Neurologico C. Besta, Via Celoria tory involvement is frequent and often is the dead cause. Muscle
11, 20100 Milano Italy; E-mail: Lamperti.c@istituto-besta.it biopsy shows typical dystrophic changes (degeneration and rege-

1381-6128/10 $55.00+.00 © 2010 Bentham Science Publishers Ltd.


Muscular Dystrophies Current Pharmaceutical Design, 2010, Vol. 16, No. 8 979

Fig. (1). Muscle fibers membrane.

neration of muscle fibres, and proliferation of fatty and connective 13] Only three patients right now has been described as associated
tissue). [1,5] Immunocytochemistry analysis Lamin and laminin, a- to mutations in the sarcolemmal membrane 1-7 integrin, on with
dystroglycan, collagen VI, and 7 integrin lead to diagnosis of the mental retardation associated with muscle weakness [6].
specific diseases. Finally, mutations in the nuclear involvement protein Lamin
These diseases can be classified into two major groups based on A/C are undoubtedly those that can rise the most variable clinical
the affected genes and the location of their expressed protein: phenotype, ranging from a congenital muscular dystrophy with
abnormalities of extracellular matrix proteins (LAMA2, COL6A1, contractures (Emery Dreifuss), to an isolated dilated cardio-
COL6A2, COL6A3), or abnormalities in the proteins involved in myopathy and a Charcot -Marie-.Toot disease type 2B1[14].
the glycosylation of  dystroglycan, (fukutin, POMGnT1, POMT1,
POMT2, FKRP, LARGE) [6-8]. The latter mechanism is intriguing Dystrophinopathies
and has aroused so great an interest in the last few years that this The dystrophin gene is the largest gene yet identified in
condition is described as an identity apart called dystroglyca- humans, identified for the first time in 1998. It is located in the
nopathy.  DG is a high glycosylated protein that forms a complex chromosome Xp21. [15] Its product, dystrophin, is postulated to be
with the membrane spannig Beta dystroglyca and conntecting the essential for the protection of the sarcolemma from the stress of
extracellular matrix to the cytoskeleton [9,10] (Fig. 1). While repeated contractions by providing an indirect link between the
initially a clear correlation between gene defect and phenotype was subsarcolemmal cytoskeletal actin and the intermediate filaments in
observed for each of these diseases, (Walker Warburg syndrome the muscle fibre with the extracellular matrix (Fig. 1). The muta-
was associated with mutations in POMT1 and POMT2, Fukuyama tions in the dystrophin gene, cause the disturbance of the reading
congenital muscular dystrophy with fukutin mutations, and Muscle frame, resulting in a severe reduction or absence of dystrophin in
Eye Brain disease associated with POMGnT1 mutations, have been the skeletal and cardiac muscle. Approximately 65% of patients
recently demonstrated that allelic mutations in each of these six with DMD have intragenic out-of-frame deletions and approxi-
genes can result in a much wider spectrum of clinical conditions mately 10% of them have duplications of one or more exons of the
[11] causing in some chase LGMD (see below). Thus, the crucial dystrophin gene. The remaining patients have point mutations or
aspect in determining the phenotypic severity is not to assess which other smaller gene rearrangements. Typically, out-of-frame
gene is primarily mutated, but how severely the mutation affects the dystrophin gene mutations lead to a severe reduction or absence of
glycosylation of ADG [12] Although the incoming new scientific dystrophin in the muscle, resulting in DMD phenotype, whereas in-
development, at the moment about 65% of congenital muscular frame mutations lead to the expression of a partly functional
dystrophies are without genetic diagnosis [13] and new entities truncated protein, resulting in the milder Becker muscular dys-
have been recently described with new different presentations [2- trophy (BMD) [16].
980 Current Pharmaceutical Design, 2010, Vol. 16, No. 8 Costanza and Moggio

Table 1. Clinical Feature and Genetic Classification of CMD. MDC: Congenital Muscular Dystrophy, WWS: Walker-Warburg
Syndrome, MEB: Muscle Eyes Brain Disease, CK: Creatine Kinase, RSMD Rigid Spine Muscular Dystrophy

Disease Protein Allelic Forms CNS Involvement Muscle/Respiratory Involvement

MDC1A Laminin-2 LGMD1A White matter changes, neuronal Muscular dystrophy, respiratory insufficiency and
migration abnormalities and nocturnal hypoventilation, peripheral neuropathy
mental retardation (seizures)

MDC1B ? No involvement Facial weakness, diaphragmatic involvement with early


respiratory failure, rigid spine, secondary laminin 2
deficiency

Rigid Spine Syndrome Selenoprotein-1 No involvement Muscular dystrophy, axial hypotonia and weakness,
(RSMD1) lumbar scoliosis and cervical spine stiffness, respiratory
failure due to skeletal abnormalities and diaphragmatic
weakness, CK normal or mildly elevated

Integrin a7 Congenital Integrin 7 Mental retardation Mild muscular dystrophy/myopathy, torticollis, mildly
Myopathy elevated CK

Ullrich's Disease Collagen VI No involvement Neonatal muscle weakness, kyphosis of spine, joint
(1,2,3) contractures, torticollis, hip dislocation,
hyperextensibility of distal joints, cheloid formation,
invariable respiratory insufficiency, normal intelligence,

MDC1D LARGE Profound mental retardation, Muscular dystrophy


brain alterations

WWS POMT1 LGMD2K Type II lissencephaly/agyria, Muscular dystrophy, CK elevated


POMT2 hydrocephalus, eye abnormalities

MEB POMGnT1 LGMD2M Eye abnormalities and abnormal Muscular dystrophy, broad clinical phenotype,
neuronal migration significant hypotonia,

Fukuyama CMD Fukutin LGMD2L Severe mental retardation, Muscular dystrophy, cardiomyopathy.
seizures, eye abnormalities

MDC1C FKRP LGMD2I Structural brain changes, retinal Muscular dystrophy, congenital weakness and
changes with blindness hypotonia, calf and tongue hypertrophy, shoulder muscle
wasting, cardiomyopathy, CK elevated

DMD is the most common form of muscular dystrophy BMD patients present symptoms similar to DMD but the
affecting one in every 3500 live male births. Almost all boys with severity of the disease is variable presenting as a severe form DMD
DMD are symptomatic before the age of 5 years, with calf like [19], a isolated dilated cardiomyopathy [20], a pseudometa-
hypertrophy, toe/waddling gait and Gowers’ sign. Subsequently, bolic form characterized by muscle cramps with myoglobinuria
they develop difficulties in running, jumping, and climbing steps. [21], a quadriceps myopathy [22]. Finally patients could be
Usually tendon retraction becomes evident in the first decade of life completely asymptomatic showing only high serum CK levels. In
associated with waddling gait, and lumbar lordosys. Independent all the forms, the clues of the diagnosis is the calf hypertrophy that
ambulation is lost between 10 and 14 years, with subsequent dete- is always present [19].
rioration in respiratory function and development of contractures The severity of the disease is usually correlated with the amount
and scoliosis [17]. A cognitive evaluation carried on in twenty of remnant functional dystrophin. Patients with DMD have less
DMD patients focused on memory, information processing/learning than 5% of the quantity of normal dystrophin. Patients with BMD
ability, and executive functions, conclude that there is a worsening have at least 20% normal dystrophin levels [23]. Most patients with
in these patients compared to age matched control in the short term BMD (85%) have dystrophin of abnormal molecular weight, often
memory and learning ability, while no significant difference was with reduced dystrophin quantity. A minority have normal-sized
evident in general intellectual ability [18]. protein of reduced quantity. Because of this correlations between
Muscular Dystrophies Current Pharmaceutical Design, 2010, Vol. 16, No. 8 981

the amount of dystrophin and the clinical features, the study of TRIM32), and the glycosylation pathway enzymes (Fukutin Related
muscle biopsy is important in these diseases, and the genetic study Protein, Protein O-Mannosyltransferase 2) [30-33].
is not enough to give prognostic information to patients and their Clinically, these muscular dystrophies present with progressive
families. muscle weakness and atrophy with predominant involvement of
DMD/BMD carrier. Females may be asymptomatic or may scapular and pelvic girdles. The neck flexors and extensors may be
have mild muscle weakness with an increased CK, calf hyper- involved and typically facial muscles are not involved and
trophy, myalgia, and cramps, and are at risk of dilated cardio- cognition is usually normal. Cardiac or other systemic involvement
myopathy [24]. Rarely, carrier female can present clinical mani- is variable and can be present in LGMD 1B, 1D, 2C-G, and 2I.
festation similar to DMD patients. Approximately 50% of DMD Typically elevated serum creatine kinase (CK) levels are present.
carrier females will have no family history of neuromuscular Nevertheless, LGMDs usually present a large clinical variability
disease [25]. The muscle biopsy could be normal or slightly patho- regarding age of onset, patterns of skeletal muscle distribution,
logical with a mild increase of connective tissues. Dystrophin heart damage, respiratory involvement and rate of progression that
immunocytochemistry in carrier or symptomatic females shows a result in variable morbidity and disability. In some cases such as
mosaic pattern, with a combination of fibres either containing or caveolinopathy, a rippling phenomena, cramps and calf hypertrophy
lacking dystrophin. Quantitative genetic studies or sequencing may are the main features [34]. Nevertheless, calf hypertrophy is com-
then be confirmatory. mon also in LGMD2I [17]. Serum CK is usually modestly elevated
Before the age of 5 years, serum CK levels are 10 to 200 times but can be very high in the sarcoglycanopathies, dysferlinopathy,
higher than normal levels, in boys with DMD and BMD, but and caveolinopathy. The autosomal recessive LGMDs generally
decline with advancing age. The CK does not discriminate between have an earlier onset, more rapid progression and higher serum CK
dystrophinopathies. Nerve conduction studies are normal. values than the dominant LGMDs. EMG shows myopathic changes
Electromyography shows myopathic changes. The muscle biopsy is and is particularly useful in discriminating mild forms of dominant
the main instrument to make diagnosis of DMD and BMD. LGMD, which may be associated with only very mild serum CK
Classically, it shows degeneration, regeneration, and variability of elevation, from spinal muscular atrophy type II and chronic
fibre size with replacement of muscle by fat and connective tissue. inflammatory neuropathies. The muscle biopsy still represents an
Milder changes are seen in BMD. Immunocytochemical studies are important step in the diagnostic process.
fundamental to differentiate DMD to BMD. In DMD a complete Muscle biopsy could range from normal biopsy to a severe
absence of binding with the antibodies for the COOH, NH2 and dystrophic pattern. In dystrophies such as dysferlynomathy and
Rod domain is observed. A partial reduction or the presence of a calpainopathy, a severe inflammation is often present and it is
truncated proteins lead to the diagnosis of BMD. The immunbolt is characterized by large infiltrates and several necrosis, while these
a more sensitive test, and it has to be used to confirm the diagnosis. aspects are really rare in caveolinopathy [30]. Moreover, in dysfer-
Genetic testing techniques include the polymerase chain reaction linopathy, several fibres in regeneration are present, indicating that
(PCR), western blotting, mutation scanning, and/or sequence ana- the dysferlin protein may be involved in the regenerating process,
lysis [17] .The DNA extracted from whole blood or isolated white by controlling cytokine expression [27]. Some vacuoles could be
cells can be used for diagnosis. Prenatal diagnosis is routinarily present in the muscle biopsy in LGMD1A. The immunostaing for
done in DMD,BMD carrier . Recently the MLPA system has been the sarcoglycan, dysferlyn, caveolin and alpha dystroglycan is
identified as useful to rapidly identify duplication and deletion in normally used for the diagnosis. Recently the immunostaing for
the dystrophin gene [26]. Pulmonary function studies, electrocar- calpain 3 has been found useful for the diagnosis in association with
diography, and echocardiography should be performed annually the immunoblot for the specific proteins [30-31]. However, protein
after 10 years old. deficiency documented by immunohistochemistry in muscle may be
secondary and in most patients a definite diagnosis can be obtained
Limb-Girdle Muscular Dystrophies only by genetic analysis, as the genetic test confirms the diagnosis.
The Limb Girdle Muscular Dystrophies (LGMD) are a hetero- Treatment is symptomatic.
geneous group of inherited progressive muscle disorders affecting
predominantly shoulder and pelvic girdle muscles. There are at General Management
least eighteen different subtypes of LGMD, seven with an auto- A specific therapy for muscular dystrophy is not available, thus
somal dominant ineritans (LGMD1A-G) and eleven with an considering the possible role of muscle inflammation in the patho-
autosomal recessive pattern of inheritance (2A-K) (Table 2) [27- genesis of the disease, the corticosteroid is the main drug used in
29]. dystrophic patients and in particular in DMD. This therapy is
AD-LGMD associated with physiotherapy and orthesis management of heart
and respiratory complications, scoliosis and nutritional aspects.
The autosomal dominant forms (LGMD1) represent probably
less than 10% of all LGMD cases. The proteins involved in AD Randomized controlled clinical trials of daily oral prednisone
LGMD are structural proteins such as myotilin, a component of the demonstrated an improvement in muscle strength and function in
sarcomeres responsible for the LGMD1A, Lamina A/C, part of the children with DMD, compared to baseline and to controls [35,36].
nuclear envelop for the LGMD1B, and Caveolin (LGMD1C), In these trials, a dose of 0.75 mg/kg/day of prednisone was used
involved in the constitution of caveole in the muscle membrane [37,38]. Long-term neuromuscular benefits of corticosteroid use
(Fig. 1). For the LGMD 1D-G all loci have been identified by include prolonged independent ambulation and a lower prevalence
linkage analysis, but the genes involved are still unknown [29]. The of scoliosis [39-41]. Adverse effects of prednisone, in particular
onset of the disease is variable ranging from infancy to 2nd-3rd related to behaviour, weight, and bone density often necessitate the
decade of life. reduction of the dose over time.
AR- LGMD The earliest sign of DMD is a progressive decline in limb
muscles function. This is primarily due to skeletal muscle weakness
All mapped genes responsible for autosomal recessive forms and secondarily to joint contractures. Physiotherapy is the primary
(LGMD2) have been identified. They encode highly different therapy to prevent joint deformities 9. In DMD, rehabilitation in
proteins involved in all aspects of muscle cell biology, such as the knee, ankle, foot orthoses (KAFOs) at the time of loss of inde-
sarcolemmal muscle membrane (sarcoglycans, dysferlin), the pendent walking is effective in prolonging walking for an average
sarcomere (telethonin, titin), the muscle cytosol (calpain-3, of 18 months to 2 years. This has been associated with reduced
incidence of scoliosis.
982 Current Pharmaceutical Design, 2010, Vol. 16, No. 8 Costanza and Moggio

Table 2. Clinical Features and Genetic Classification of LGMD. LGMD: Limb Girdle Muscular Dystrophy, AR: Autosomal
Recessive, AD: Autosomal Dominant, AO: Age of Onset, IH, Immunohistochemistry DG: Dystroglycan

LGMD HE Protein AO Clinical Manifestations Muscle Biopsies Cardiac Involvement

LMGD 1A AD Myotilin II-III Dysfagia and myopathy Dystrophic changes


Vacuoles, accumulation of myotilin at IH

LMGD 1B AD Laminin A/C I-III Myopathy, CMT2, congenital Dystrophic changes Common
myopathy.

LMGD 1C AD Caveolin 3 I-IV Calf hypertrophy, Rippling, High variability in fibers size, absence of Un common
muscle weakness caveolin at IH

LMGD 2A AR Calpain 3 I-IV Posterior compartments of the Dystrophic changes, regeneration, Un common
limbs reduction of calpain 3 at WB

LMGD 2B AR Dysferlin I-IV Distal myopathy. Dystrophic changes , inflammatory cells , Un common
reduction of dysferlin at WB

LMGD 2C-D- AR Sarcoglycan  I-II DMB like, BMD Like, calf Dystrophic changes, reduction of one or Common
E-F hypertrophy all sarcoglycans at IH

LMGD 2G AR Telethonin I-II Calf hypertrophy or hypotrophy, Dystrophic changes, reduction of Un common
distal leg weakness Telethonin at IH

LMGD 2H AR TRIM32 I-IV Possible mild facial weakness Dystrophic changes

LMGD 2I AR FKRP I-IV Calf hypotrophy, myopathy, Dystrophic changes and reduction of  Common
muscle pain DG

LGMD 2J AR Titin I-II Distal weakness, proximal distal Dystrophic changes Common
myopathy

Respiratory and cardiac care are two aspects that have to be especially during intense contractile activity. This results in an
monitored. In the past, the onset of symptomatic sleep hypoven- increased calcium entry and focal or diffuse damage to the fibre
tilation signified imminent demise, as the only way to prolong life [51]. Damaged or dead fibres can be repaired or replaced by
was mechanical ventilation through tracheotomy. The greatest satellite cells [52]. These cells are actually considered the resident
contribution to improve the respiratory care, leading to extend the ‘stem-like’ cells in skeletal muscle. They are responsible for muscle
longevity in DMD, is the domiciliary non-invasive ventilation in growth and regeneration in postnatal life [53]. However, dystrophic
1990 [42-44]. Moreover, it seems that long term steroid therapy satellite cells share the same molecular defect and produce fibres
would improve pulmonary function. that are also prone to degeneration. With time, the population of
Dilated cardiomyopathy is sometimes associated with muscular satellite cells is exhausted and the muscle tissue is progressively
dystrophy causing ventricular dysfunction, defined as a shortening replaced by connective and adipose tissue. Moreover, the death of
fraction, less than 28%. Nevertheless, corticosteroid treatment is muscle cells provokes a secondary inflammatory reaction inducing
protective against this dysfunction, if it starts prior to cardiac a further muscle damage.
involvement [45,46]. Thus, as said before, there is not a cure for these diseases,
Early angiotensin-converting enzyme inhibitor (ACEi) treat- therefore there is a great interest in developing new promising
ment may also be indicated in DMD as suggested by a trial of strategies and therapies. One of these strategies consists in the
perindopriol, although some cardiologists do not find any treatment tentative restoration of the normal proteins with the aim of
necessary for complication that are often asymptomatic for a long correcting the gene defect and the regenerations of muscle tissues
time [47]. There are prospective trials of ACEi and angiotensin II- using stem cells or factors that promote regenerations. Each stra-
type 1 receptor blockers in patients with dilated cardiomyopathy tegy has both advantages and limitations: for example, strategies
from a variety of aetiologies, indicating improvement both in that repair the dystrophin gene might be suitable only for a subset
survival and in symptoms [48-50]. Such studies argue for early and of mutations, whereas cell and gene therapies are limited by costs
continued use of corticosteroids and ACEi/angiotensin II-type 1 related to cell or vector production, and they can be available only
receptor blockers in patients with DMD. for a limited number of patients [54]. These strategies are: 1 gene
therapy, 2 cell therapy, 3 drug therapy
NEW PROSPECTIVE IN THERAPY
Gene Therapy
Muscle weakness in muscular dystrophy is secondary to muscle
degeneration due to a mutations in genes composing the DGC Exon Skipping
(dystrophyn glycoprotein complex). Mutation in one of these genes As previously described, muscular dystrophies are secondary to
affects proteins that form a link between the cytoskeleton and the mutations in genes encoding for proteins of the DGC. In particular,
basal lamina. Absence of one of these proteins often causes the it is well known that either the deletions or the mutations in the
disassembly of the whole multiprotein complex associated with dystrophin gene produce an alteration of the reading frame of the
dystrophin, leading to increased fragility of the sarcolemma, gene. One therapeutic strategy is the use of an antisense oligo-
Muscular Dystrophies Current Pharmaceutical Design, 2010, Vol. 16, No. 8 983

nucleotide (OAs) to modify the dystrophin mRNA splicing. The secondary outcome is the measurement of the mini-dystrophin gene
OAs prevent the normal splicing of the gene, by masking crucial expression at the site of gene transfer and the muscle strength
areas of the messenger RNA during the splicing process that evaluation by Maximal Volume Isometric Contraction Testing.
produce an “exon skipping “. The skipping leads to the restoration
of the reading frame of the mRNA and that produces (producing) a Cell Therapy
phonotype similar to the milder BMD and provides significant Cell-based therapy is a potential solution toward restoring
clinical improvement in DMD patients. It has been estimated that dystrophin expression in skeletal muscles. This therapy exploits the
the skipping of 12 exons around exon 51 could be therapeutic for ability of wild-type muscle precursor cells to fuse with damaged
about 20 % of dystrophin deletions [55]. An open trial on four DMD myofibers, thereby introducing nuclei that express the normal
patients affected with DMD using a 2-O-methyl phosphorothioate dystrophin gene in the muscle syncytia. Several adult stem cell have
antisense oligonucleotide, demonstrated the skipping of exon 51 been isolated, and are characterized and used in animal trans-
and the sarcolemmal dystrophin expression 28 days after single plantation experiments [64]. A few examples of stem cells showing
injection into the tibialis anterior muscle [56]. The amount of a potential therapeutic affecting or used in animal clinical trial are
dystrophin produced ranged from 3% to 12% of control specimens listed below.
by Western blot and from 17% to 35% of control specimens by Muscle Derived Stem Cell (MDSC)
ratio of dystrophin to laminin _2 considering the immunohisto-
chemistry enhance expression [56]. Recently, a single-blind Muscle satellite cells play a central role in postnatal muscle
placebo-controlled dose-escalation study in seven patients with growth and regeneration, and are promising tool for cell therapy.
DMD, was carried out to assess the safety and biochemical efficacy Satellite cells are dormant progenitors located at the periphery of
of an intramuscular morpholino splice-switching oligonucleotide skeletal myofibers that can be triggered to proliferate for both self-
(AVI-4658) that skips exon 51 in dystrophin mRNA. The intra- renewal and differentiation into myogenic cells secondary to
muscular injection in the extensor digitorum brevis (EDB), and the specific stimuli such as oxidative stress [65] were able to directly
contralateral muscle have been taken as a control. This trial showed isolate a pure population of satellite cells, injecting them into
that the intramuscular AVI-4658 was safe, and induced the local dystrophic dogs and restoring dystrophin expression 3 weeks post-
expression of dystrophin within treated muscles. This proof-of- transplantation. Into irradiated dystrophic mice, they also formed a
concept study could be the starting point to a systemic clinical trial small amount of the satellite cell pool that expressed both Pax7 and
[57]. Pax3 [65]. The results obtained in the mouse model led to test the
myoblast injection in DMD patients in phase I clinical trials.
Nonsense Suppression Unfortunately, these trials demonstrated that myoblast transplan-
About the 15% of DMD mutations are stoop codon point muta- tation is an inefficient technique: the efficiency of the dystrophin
tions that produce a truncated or absent protein. Aminoglycosides production in muscle fibres of DMD patients was very low ( 1%)
cause misreading of the RNA code at the premature but not at the and there was no functional or clinical improvement in the children
normal termination codons, leading to the insertion of alternative [66].
amino acids at the site of the mutated codon, transcription and Mesoanguiblast
protein formation. Gentamicin has been found to be effective in
mdx mice in some cases [58], whereas it is ineffective in other Mesoangioblasts in mice and dogs and pericyte in human are
cases, (it is ineffective) [59]. Moreover, gentamicin presents a lot of multipotent progenitors of mesodermal tissues, physically
side effect in long term administration. A similar molecule, associated with the embryonic dorsal aorta in avian and mammalian
PTC124, is now available. This molecule specifically allows to species. Cossu, et al in 2003 demonstrated the capacity of
read-trough the nonsense mutations, without any alteration on mesoangioblasts to differentiate in various mesodermal phenotypes
normal translational stop signs. PTC 124 was well tolerated in qualifying these progenitors as a novel class of stem cells. Systemic
phase 1 studies and in 62 healthy volunteers [60]. A phase 2A open delivery in dystrophic mice and dogs has been performed [67]. The
label trial is currently ongoing, and will evaluate both muscle intra-arterial transplantation of donor mesoangioblasts ameliorated
strength and dystrophin expression, and serum CK levels. A defective muscle structure and function in dystrophic mice [68] and
randomized, controlled 48-week treatment phase 2B trial in DMD in cyclosporine immunosoppressed dogs [69]. In particular, Cossu,
and BMD patients with nonsense mutations has begun, being the et al. showed that dogs treated with mesoangioblasts had an
total distance covered during a 6-minute walking test the primary extensive expression of dystrophin in the majority of the muscle
outcome measure (NCT00592553, clinicaltrials.gov). This study fibres and were characterised by both normal force contraction and
has been recently extended to other 96 weeks. amelioration of defective mobility [69]. Recently, similar results
were achieved with mesoangioblasts transplanted into mdx/utrophin
AVV Mediated Gene Transfer null mice [70]. Because of these encouraging results, a clinical trial,
The third therapeutic possibility to restore the dystrophin in the that uses donor stem cells from an HLA (human leukocyte antigen)
muscle plasma membrane is represented by the adenoviral vector identical donor have been planed. However, this study, at least in
(AVV). The AVV is the leading viral vector for human gene dogs, do not provide control animals treated only with cyclosporin
therapy, but it has a maximum package of 5 kb, while the dys- A (CSA) [70,71]. The possible beneficial effect of CSA in muscular
trophin gene is about 11 kb. For this reason, a truncated dystrophin dystrophy is currently being tested in a specific clinical trial in
gene construction has been provided, and it is called mini or micro- Germany [72]. Nevertheless, one paper reports beneficial effects of
dystrophin. Mini-dystrophins and micro-dystrophins have been the drug in dystrophic mice whereas other papers clearly show a
shown to ameliorate the dystrophic pathology of the mdx mouse deleterious long-term effect, resulting in the inhibition by CSA of
[61-63]. This strategy allows to express a truncated but partly the calcineurin pathway that is essential for muscle regeneration
effective dystrophin protein in muscle, thus the adminis-tration of a [73-75]. A study in dogs involving a restricted number of animals
viral agent could produces an immune response to the vector. A showed a great variability in the progression of the disease. For all
phase I trial of a micro-dystrophin under a cytomegalo-virus these reasons, a great prudence has to be used before treating
promoter in a modified AAV is now underway in boys with DMD patients. No adverse events related to mesoangioblast infusion have
(NCT00428935, clinicaltrials. gov). A phase I study has started been observed in a total of 16 dogs treated [69] or currently under
now and will provide useful information about safety, and the treatment, and a group in Italy is going to perform a clinical trial in
DMD patients.
984 Current Pharmaceutical Design, 2010, Vol. 16, No. 8 Costanza and Moggio

Circulating Stam Cell myostatin inhibition by follistatin, which opposes the activity of
Several studies have demonstrated that wild-type (wt) total myostatin mentioned above, is a good method to improve myoblast
bone marrow-derived or side population (BM-SP) cells are transplantation and muscle function. [90].
incorporated into regenerating skeletal muscle fibres when TGF-1
transplanted into dystrophic mice [76-78]. However, in some cases TGF-B1 is another member of the TGF-  superfamily. It is
transplanted cells failed to restore the expression of that protein, homologous to myostatin, and induces many similar effects on
suggesting that under standard conditions they have little muscle, including the inhibition of proliferation, differentiation of
therapeutic potencial [79,80]. In other cases, results have been more muscle precursor cells and stimulating the fibrosis [91-93].
encouraging. For example, bone marrow mesenchymal stem cells, Contrary to myostatin, TGF-1 is up-regulated in early DMD [94-
which show little myogenic differentiation, became highly 96]. These features would suggest that the inhibition of TGF-  1
myogenic when engineered to express high levels of intracellular would be a possible therapeutic strategy. However, TGF  1 is
Notch protein, and produced dystrophin in many fibres when widely expressed and controls a range of processes in various
transplanted into mdx mice [77]. Another cell population, isolated tissues throughout the body, raising concerns of specificity and
from blood and from skeletal muscl expressing the stem cell CD133 toxicity. Thus, some studies have showed that losartan, a
antigen, has been shown to give rise to dystrophin- positive fibers angiotensin II-type1 receptor bloker usually used for hypertension,
when transplanted into scid (Severe Combined Immuno- is well tolerated in mdx mice mdx mice treated for 6 to 9 months
deficiency)/mdx mice [76]. Torrente et colleagues, demonstrated with this drug demonstrated less fibre size heterogeneity, less
that this cells can be injected safely in DMD patients not only fibrosis per area, and increased strength by grip strength and
without side effects but also promoting an increase in the number of absolute force of explanted muscle [97]. Many older DMD patients
capillaries per muscle fibres [81], and that DMD CD133+ cells can are currently being treated with losartan for the presumptive cardiac
be genetically modified to re-express a functional dystrophy [82]. benefits of angiotensin II-type 1 receptor blockers.
Future clinical trials with this cell types are now feasible.
CONCLUSION
Drug Theraphy
Muscular dystrophies are severe progressive muscle diseases
The last strategy is to stimulate growth and regeneration of involving both adults and children, characterized by the degene-
dystrophic muscle or to inhibit the inflammation by promoting ration of muscle tissues and substitution of muscle fibres with
muscle degenerations. These strategies may provide functional connective tissues. The supporting therapy such as corticosteroids,
benefits by increasing size and strength of minimally affected phisiokinesitherapy, hart and non invasive ventilatory support are
muscle, or by improving the quality of the composition of useful tools to assist these patients and arise the possibility of a
dystrophic muscle. Thus, they will not be a cure for the diseases, in better quality of life and a longer life expectancy for these patients.
fact they do not address the pathophysiology of the single disease, Although a therapy for these diseases has not been identified,
such as the loss of dystrophin or sarcoglycans, but it can produce multiple novel therapeutic agents have recently entered clinical
benefits in all the dystrophic muscles, as it is focused on the trials or are expected to in the near future. The therapy strategies
pathomechanism of the dystrophic changes. Postnatal growth and involve gene expression modulations, cell therapy and the
regeneration are regulated by a variety of endogenous growth stimulation of regenerating factor. It is unlike that any of the current
factors, such as members of the transforming growth factor-. approaches will independently cure this devastating disease, but it
Myostatin is possible that the combination of more agents could provide a
Myostatin is a member of the TGF-  superfamily and is an therapeutic approach. Nevertheless, great part of the pathogenesis
endogenous negative regulator of muscle growth. [83]. Loss of and the pathomechanism of these diseases remains to be under-
function mutations in myostatin results in massive muscle stood. Even if, since 1993, 600 papers about exon skipping and 140
hypertrophy and hyperplasia with approximately doubling of papers in stem cells were published, and right now there are
muscle mass in humans, as described in one patient with a loss of numerous trial in animals and humans being performed all the
function mutation in the myostatin gene [84,85]. Because myostatin world, a useful therapy is still unknown. In our opinion the effective
is a negative regulator of muscle mass, several studies have been pharmacological agent or a combination of strategies might be
performed to investigate a potential therapeutic effect of antago- found when more will be understood about the pathophysiology of
nizing the myostatin pathway. Myostatin can be inhibited post-natal these disorders.
by a variety of mechanisms that similarly induce muscle growth
REFERNCES
[86]. Inhibition of myostatin with a neutralizing antibody and with a
modified myostatin pro-peptide, delivered via AAV, ameliorates References 99-101 are related articles recently published.
the dystrophic phenotype of the mdx mouse model [87,88]. Based
[1] Muntoni F, Voit TC. The congenital muscular dystrophies in 2004:
on these results Wyeth Pharmaceuticals (http: //www. wyeth.com/) a century of exciting progress. Neuromuscul Disord 2004; 14: 635-
developed neutralizing antibodies among which MYO-029 has 49.
entered in clinical trials for different forms of muscular dystrophy, [2] Fagiolari G, Cappellini A, Cagliani R, Prelle A, Lucchini V,
in particular BMD. No severe adverse events were observed in the Fortunato F, et al. Muscular dystrophy: central nervous system -
short term treatment except hypersensitivity to the antibody in some dystroglycan glycosylation defects and brain malformation. J Child
patients. The first clinical trial of a myostatin inhibitor in a disease Neurol 2009; in press.
population was recently completed demons-trating safety without [3] Bertini E, Pepe G. Collagen type VI and related disorders: bethlem
target-related side effects of a neutralizing antibody to myostatin, myopathy and Ullrich scleroatonic muscular dystrophy. Eur J
MYO-029 in subjects with diverse adult muscular dystrophies, Paediatr Neurol 2002; 6(4): 193-8.
[4] Quijano-Roy S, Renault F, Romero N, Guicheney P, Fardeau M,
including Becker muscular dystrophy [89]. Long-term safety and Estournet B. EMG and nerve conduction studies in children with
efficacy are currently being tested. Further information is available congenital muscular dystrophy. Muscle Nerve 2004; 29: 292-9.
on the website of the Muscular Dystrophy Association (http: [5] Jones K, North K. The congenital muscular dystrophies. In: Jones
//www.mdausa.org). Improvement in muscle strength and function HR, De Vivo DC, Darras BT, Eds. Neuromuscular Disorders of
were not demonstrated in this trial, while biological activity of Infancy, Childhood, and Adolescence: A Clinician’s Approach.
MYO-029 was suggested by trends in increased muscle mass and Philadelphia: Butterworth Heinemann 2003; pp. 633-644.
fibres diameters [89]. In the future, this therapy could be combined
with gene or cell therapy. Recently, some authors demonstrated that
Muscular Dystrophies Current Pharmaceutical Design, 2010, Vol. 16, No. 8 985

[6] Dubowitz V, Fardeau M. Proceedings of the 27th ENMC spon- Italian limb girdle muscular dystrophy patients. Hum Mutat 2008;
sored workshop on congenital muscular dystrophy. Neuromuscul 29(2): 258-66.
Disord 1995; 5: 253-8. [31] Guglieri G, Straub V, Bushby K, Lochmüller H. Limb-girdle mus-
[7] van Reeuwijk J, Janssen M, van den Elzen C, Beltran-Valero de cular dystrophies. Curr Opin Neurol 2008; 21: 576-84.
Bernabé D, Sabatelli P, Merlini L, et al. POMT2 mutations cause [32] Clement EM, Godfrey C, Tan J, Brockington M, Torelli S, Feng L,
-dystroglycan hypoglycosylation and Walker Warburg syndrome. et al. Mild POMGnT1 mutations underlie a novel limb-girdle
J Med Genet 2005; 42(12): 907-12. muscular dystrophy variant. Arch Neurol 2008; 65: 137-41.
[8] Longman C, Brockington M, Torelli S, Jimenez-Mallebrera C, [33] Biancheri R, Falace A, Tessa A, Pedemonte M, Scapolan S,
Kennedy C, Khalil N, et al. Mutations in the human LARGE gene Cassandrini D, et al. POMT2 gene mutation in limb-girdle
cause MDC1D, a novel form of congenital muscular dystrophy muscular dystrophy with inflammatory changes. Biochem Biophys
with severe mental retardation and abnormal glycosylation of alpha Res Commun 2007; 363: 1033-7.
dystroglycan. Hum Mol Genet 2003; 12(21): 2853-61. [34] Godfrey C, Clement E, Mein R, Brockington M, Smith J, Talim B,
[9] Barresi R, Campbell KP. Dystroglycan: from biosynthesis to et al. Refining genotype phenotype correlations in muscular
pathogenesis of human disease. J Cell Sci 2006; 119(Pt 2): 199- dystrophies with defective glycosylation of dystroglycan. Brain
207. 2007; 130: 2725-35.
[10] Cohn RD. Dystroglycan: important player in skeletal muscle and [35] Griggs RC, Moxley RT, 3rd, Mendell JR, Fenichel GM, Brooke
beyond. Neuromuscul Disord 2005; 15(3): 207-17. MH, Pestronk A, et al. Prednisone in Duchenne dystrophy: a
[11] Muntoni F, Brockington M, Blake JD, Torelli S, Brown SC. randomized, controlled trial defining the time course and dose
Defective glycosylation in muscular dystrophy. Lancet 2002; response. Clinical Investigation of Duchenne Dystrophy Group.
360(9343): 1419-21. Arch Neurol 1991; 48: 383-8.
[12] Muntoni F, Brockington M, Godfrey C, Ackroyd M, Robb S, [36] Mendell JR, Moxley RT, Griggs RC, Brooke MH, Fenichel GM,
Manzur A, et al. Muscular dystrophies due to defective Miller JP, et al. Randomized, doubleblind six-month trial of
glycosylation of dystroglycan. Acta Myol 2007; 26(3): 129-35. prednisone in Duchenne’s muscular dystrophy. New Engl J Med
[13] Mercuri E, Messina S, Bruno C, Mora M, Pegoraro E, Comi GP, 1989; 320: 1592-7.
et al. Congenital muscular dystrophies with defective glycosylation [37] Moxley RT, III, Ashwal S, Pandya S, Connolly A, Florence J,
of dystroglycan: a population study. Neurology 2009; 72(21): Mathews K, et al. Practice parameter: corticosteroid treatment of
1802-9. Duchenne dystrophy: report of the Quality Standards Subcom-
[14] Rankin J, Ellard S. The laminopathies: a clinical review. Clin mittee of the American Academy of Neurology and the Practice
Genet 2006; 70(4): 261-74. Review. Erratum in: Clin Genet 2007; Committee of the Child Neurology Society. Neurology 2005; 64:
71(3): 293. 13-20
[15] Darras BT. Molecular genetics of Duchenne and Becker muscular [38] Manzur AY, Kukntzer T, Pike M, Swan A. Glucocorticoid
dystrophy. J Pediatr 1990; 117: 1-15. corticosteroids for Duchenne muscular dystrophy (Review).
[16] Manzur AY, Muntoni F. Diagnosis and new treatments in muscular Cochrane Libr 2006: 1-41.
dystrophies. J Neurol Neurosurg Psychiatry 2009; 80(7): 706-14. [39] Alman BA, Raza SN, Biggar WD. Steroid treatment and the
[17] Darras BT. Muscular dystrophies. Continuum 2006; 12: 33-75. development of scoliosis in males with duchenne muscular
[18] Wicksell RK, Kihlgren M, Melin L, Eeg-Olofsson O. Specific dystrophy. J Bone Joint Surg Am 2004; 86-A: 519-24.
cognitive deficits are common in children with Duchenne muscular [40] Biggar WD, Harris VA, Eliasoph L, Alman B. Long-term benefits
dystrophy. Dev Med Child Neurol 2004; 46: 154-9. of deflazacort treatment for boys with Duchenne muscular
[19] Bushby KM, Gardner-Medwin D. The clinical, genetic and dystrophy in their second decade. Neuromuscl Disord 2006; 16:
dystrophin characteristics of Becker muscular dystrophy: I. Natural 249-55.
history. J Neurol 1993; 240: 98-104. [41] King WM, Ruttencutter R, Nagaraja HN, Matkovic V, Landoll J,
[20] Gedeon AK, Wilson MJ, Colley AC, Sillence DO, Mulley JC. X Hoyle C, et al. Orthopedic outcomes of long-term daily
linked fatal infantile cardiomyopathy maps to Xq28 and is possibly corticosteroid treatment in Duchenne muscular dystrophy.
allelic to Barth syndrome. J Med Genet 1995; 32: 383-8. Neurology 2007; 68: 1607-13.
[21] Gospe SM, Lazaro RP, Lava NS, Grootscholten PM, Scott MO, [42] Eagle M, Baudouin S, Chandler C, Giddings DR, Bullock R,
Fischbeck KH. Familial X-linkedmyalgia and cramps: a Bushby K, et al. Survival in Duchenne muscular dystrophy:
nonprogressive myopathy associated with a deletion in the improvements in life expectancy since 1967and the impact of home
dystrophin gene. Neurology 1989; 39: 1277-80. nocturnal ventilation. Neuromuscl Disord 2002; 12: 926-9.
[22] Sunohara N, Arahata K, Hoffman EP, Yamada H, Nishimiya J, [43] Mohr CH, Hill NS. Long-term follow-up of nocturnal ventilatory
Arikawa E, et al. Quadricepsmyopathy: forme fruste of Becker assistance in patients with respiratory failure due to Duchennetype
muscular dystrophy. Ann Neurol 1990; 28: 634-9. muscular dystrophy. Chest 1990; 97: 91-6.
[23] Hoffman EP, Kunkel LM. Dystrophin abnormalities in [44] Simonds AK, Muntoni F, Heather S, Fielding S. Impact of nasal
Duchenne/Becker muscular dystrophy. Neuron 1989; 1019-29. ventilation on survival in ypercapnic Duchenne muscular
[24] Hoogerwaard EM, van der Wouw PA, Wilde AA, Bakker E, Ippel dystrophy. Thorax 1998; 53: 949 -52.
PF, Oosterwijk JC, et al. Cardiac involvement in carriers of [45] Wagner KR. Approaching a new age in duchenne muscular
Duchenne and Becker muscular dystrophy. Neuromuscul Disord dystrophy treatment. Neurotherapeutics 2008; 5(4): 583-91.
1999; 9: 347-51. [46] Markham LW, Kinnett K, Wong BL, Woodrow Benson D, Cripe
[25] Hoffman EP, Arahata K, Minetti C, Bonilla E, Rowland LP. LH. Corticosteroid treatment retards development of ventricular
Dystrophinopathy in isolated cases of myopathy in females. dysfunction in Duchenne muscular dystrophy. Neuromuscl Disord
Neurology 1992; 42: 967-75. 2008; 18: 365-70.
[26] Pikó H, Vancsó V, Nagy B, Bán Z, Herczegfalvi A, Karcagi V. [47] Duboc D, Meune C, Lerebours G, Devaux JY, Vaksmann G,
Dystrophin gene analysis in Hungarian Duchenne/Becker muscular Bécane HM. Effect of perindopril on the onset and progression of
dystrophy families - detection of carrier status in status in left ventricular dysfunction in Duchenne muscular dystrophy. J Am
symptomatic and asymptomatic female relatives. Neuromuscul Coll Cardiol 2005; 45: 855-7.
Disord 2009; 19(2): 108-12. [48] The CONSENSUS Trial Study Group. Effects of enalapril on
[27] Bushby KMD. The limb-girdle muscular dystrophies - multiple mortality in severe congestive heart failure. Results of the
genes, multiple mechanisms. Hum Mol Genet 1999; 8: 1875-82. Cooperative North Scandinavian Enalapril Survival Study
[28] D'Amico A, Tessa A, Bruno C, Petrini S, Biancheri R, Pane M, (CONSENSUS). New Engl J Med 1987; 316: 1429-35.
et al. Expanding the clinical spectrum of POMT1 phenotype. [49] The SOLVD Investigators. Effect of enalapril on survival in
Neurology 2006; 66: 1564-7. patients with reduced left ventricular ejection fractions and
[29] Guglieri M, Magri F, Comi GP. Molecular etiopathogenesis of limb congestive heart failure. New Engl J Med 1991; 325: 293-302.
girdle muscular and congenital muscular dystrophies: boundaries [50] Pitt B, Poole-Wilson PA, Segal R, Martinez FA, Dickstein K,
and contiguities. Clin Chim Acta 2005; 361: 54-79. Camm AJ, et al. Effect of losartan compared with captopril on
[30] Guglieri M, Magri F, D'Angelo MG. Clinical, molecular, and mortality in patients with symptomaticheart failure: randomised
protein correlations in a large sample of genetically diagnosed trial—the Losartan Heart Failure Survival Study ELITE II. Lancet
2000; 355: 1582-87.
986 Current Pharmaceutical Design, 2010, Vol. 16, No. 8 Costanza and Moggio

[51] Blake DJ, Weir A, Newey SE, Davies KE. Function and genetics of [73] Schessl J. Clinical trial immunosuppressive treatment in Duchenne
dystrophin and dystrophin-related proteins in muscle. Physiol Rev muscular dystrophy - update. Neuropediatrics 2005; DOI:10.
2002; 82: 291-329. 1055/s-2005-867995 (http: //www.thieme-connect.com/ejournals)
[52] Mauro A. Satellite cell of skeletal muscle fibers. J Biophys [74] Stupka N, Gregorevic P, Plant DR, Lynch GS. The calcineurin
Biochem Cytol 1961; 9: 493-5. signal transduction pathway is essential for successful muscle
[53] Holterman CE, Rudnicki MA. Molecular regulation of satellite cell regeneration in the mdx dystrophic mice. Acta Neuropathol 2004;
function. Semin Cell Dev Biol 2005; 16: 575-84. 107: 299-310.
[54] Muntoni F, Wells D. Genetic treatments in muscular dystrophies. [75] St-Pierre SJG, Chakkalakal JV, Kolodziejczyk SM, Knudson JC,
Curr Opin Neurol 2007; 20: 590-4. Jasmin BJ, Megeney LA. Glucocorticoid treatment alleviates
[55] Cossu G, Sampaolesi M. New therapies for Duchenne muscular dystrophic myofiber pathology by activation of the calcineurin/
dystrophy: challenges, prospects and clinical trials. Trends Mol NFAT pathway. FASEB J 2004; 18: 1937-9.
Med 2007; 13(12): 520-6. [76] Parsons SA, Millay DP, Sargent MA, Naya FJ, McNally EM,
[56] van Deutekom JC, Janson AA, Ginjaar IB, Frankhuizen WS, Sweeney HL, et al. Genetic disruption of calcineurin improves
Aartsma-Rus A, Bremmer-Bout M, et al. Local dystrophin skeletal muscle pathology and cardiac disease in a mouse model of
restoration with antisense oligonucleotide PRO051. New Engl J limb-girdle muscular dystrophy. J Biol Chem 2007; 282: 10068-78.
Med 2007; 357: 2677-86. [77] Gussoni E, Soneoka Y, Strickland CD, Buzney EA, Khan MK,
[57] Muntoni F. Local restoration of dystrophin expression with the Flint AF, et al. Dystrophin expression in the mdx mouse restored
morpholino oligomer AVI-4658 in Duchenne muscular dystrophy: by stem cell transplantation. Nature 1999; 401: 390-4.
a single-blind, placebo-controlled, dose-escalation, proof-of- [78] Dezawa M. al Bone marrow stromal cells generate muscle cells and
concept study. Lancet Neurol 2009; 8(10): 918-28. repair muscle degeneration. Science 2005; 309: 314-7.
[58] Barton-Davis ER, Cordier L, Shoturma DI, Leland SE, Sweeney [79] Bachrach E, Li S, Perez AL, Schienda J, Liadaki K, Volinski J,
HL. Aminoglycoside antibiotics restore dystrophin function to et al. Systemic delivery of human microdystrophin to regenerating
skeletal muscles of mdx mice. J Clin Invest 1999; 104: 375-81. mouse dystrophic muscle by muscle progenitor cells. Proc Natl
[59] Dunant P, Walter MC, Karpati G, Lochmuller H. Gentamicin fails Acad Sci USA 2004; 101: 3581-6.
to increase dystrophin expression in dystrophin-deficient muscle. [80] Dell’Agnola C, Wang Z, Storb R, Tapscott SJ, Kuhr CS, Hauschka
Muscle Nerve 2003; 27: 624-7. SD, et al. Hematopoietic stem cell transplantation does not restore
[60] Hirawat S, Welch EM, Elfring GL, Northcutt VJ, Paushkin S, dystrophin expression in Duchenne muscular dystrophy dogs.
Hwang S, et al. Safety, tolerability and pharmacokinetics of Blood 2004; 104: 4311-8.
PTC124, a nonaminoglycoside nonsense mutation suppressor, [81] Lapidos KA, Chen YE, Earley JU, Heydemann A, Huber JM,
following single- and multiple-dose administrationto healthy male Chien M, et al. Transplanted hematopoietic stem cells demonstrate
and female adult volunteers. J Clin Pharmacol 2007; 47: 430-44. impaired sarcoglycan expression after engraftment into cardiac and
[61] Wang B, Li J, Xiao X. Adeno-associated virus vector carrying skeletal muscle. J Clin Invest 2004; 114, 1577-85.
human minidystrophin genes effectively ameliorates muscular [82] Torrente Y, Belicchi M, Marchesi C, Dantona G, Cogiamanian F,
dystrophy in mdx mouse model. Proc Natl Acad Sci USA 2000; 95: Pisati F, et al. Autologous transplantation of muscle-derived
10158 -63. CD133+ stem cells in Duchenne muscle patients. Cell Transplant
[62] Fabb SA, Wells DJ, Serpente P, Dickson G. Adeno-associated 2007; 16: 563-77.
virus vector gene transfer and sarcolemmal expression of a 144 [83] Benchaouir R, Meregalli M, Farini A, D'Antona G, Belicchi M,
kDa micodystrophin effectively restores the dystrophin-associated Goyenvalle A, et al. Restoration of human dystrophin following
protein complex and inhibits myofibre degeneration in nude/ mdx transplantation of exon-skipping-engineered DMD patient stem
mice. Hum Mol Genet 2002; 11: 733-41. cells into dystrophic mice. Cell Stem Cell 2007; 1(6): 646-57.
[63] Liu M, Yue Y, Harper SQ, Grange RW, Chamberlain JS, Duan D. [84] McPherron AC, Lawler AM, Lee SJ. Regulation of skeletal muscle
Adeno-associated virus-mediated microdystrophin expression mass in mice by a new TGF- superfamily member. Nature 1997;
protects young mdx muscle from contraction-induced injury. Mol 387: 83-90.
Ther 2005; 11: 245-56. [85] Lee S-J. Sprinting without myostatin: a genetic determinant of
[64] Farini A, Razini P, Erratico S, Torrente Y, Meregalli M. Cell based athletic prowess. Trends Genet 2007; 23: 475-7.
therapy for duchenne muscular dystrophy. J Cell Physiol 2009; [86] Schuelke M, Wagner KR, Stolz LE, Hübner C, Riebel T, Kömen
221(3): 526-34. W, et al. Myostatin mutation associated with gross muscle
[65] Montarras D, Morgan J, Collins C, Relaix F, Zaffran S, Cumano A, hypertrophy in a child. New Engl J Med 2004; 350: 2682-8.
et al. Direct isolation of satellite cells for skeletal muscle [87] Lee SJ. Regulation of muscle mass by myostatin. Annu Rev Cell
regeneration. Science 2005; 23; 309(5743): 2064-7. Dev Biol 2004; 20: 61-86.
[66] Péault B, Rudnicki M, Torrente Y, Cossu G, Tremblay JP, [88] Bogdanovich S, Krag TO, Barton ER, Morris LD, Whittemore LA,
Partridge T, et al. Stem and progenitor cells in skeletal muscle Ahima RS, et al. Functional improvement of dystrophic muscle by
development, maintenance, and therapy. Mol Ther 2007; 15: 867- myostatin blockade. Nature 2002; 420: 418-21.
77. [89] Qiao C, Li J, Jiang J, Zhu X, Wang B, Li J, et al. Myostatin
[67] Minasi MG, Riminucci M, De Angelis L, Borello U, Berarducci B, propeptide gene delivery by adeno-associated virus serotype 8
Innocenzi A, et al. The meso-angioblast: a multipotent, vectors enhances muscle growth and ameliorates dystrophic
selfrenewing cell that originates from the dorsal aorta and phenotypes in mdx mice. Hum Gene Ther 2008; 19: 241-54.
differentiates into most mesodermal tissues. Development 2002; [90] Wagner KR, Fleckenstein JL, Amato AA, Barohn RJ, Bushby K,
129: 2773-83. Escolar DM, et al. A phase I/II trial of MYO-029 in adult subjects
[68] Sampaolesi M, Torrente Y, Innocenzi A, Tonlorenzi R, D’Antona with muscular dystrophy. Ann Neurol 2008; 63: 561-71.
G, Pellegrino MA, et al. Cell therapy of alpha sarcoglycan null [91] Benabdallah BF, Bouchentouf M, Rousseau J, Bigey P, Michaud
dystrophic mice through intra-arterial delivery of mesoangioblasts. A, Chapdelaine P, et al Inhibiting myostatin with follistatin
Science 2003; 301: 487-92. improves the success of myoblast transplantation in dystrophic
[69] Sampaolesi M, Blot S, D'Antona G, Granger N, Tonlorenzi R, mice. Cell Transplant 2008; 17(3): 337-50.
Innocenzi A, et al. Mesoangioblast stem cells ameliorate muscle [92] Allen RE, Boxhorn LK. Inhibition of skeletal muscle satellite cell
function in dystrophic dogs. Nature 2006; 444: 574-9. differentiation by transforming growth factor-. J Cell Physiol
[70] Berry SE, Liu J, Chaney EJ, Kaufman SJ. Multipotential 1987; 1987: 576-2.
mesoangioblast stem cell therapy in the mdx/utrn_/_ mouse model [93] Li Y, Foster W, Deasy BM, Chan Y, Prisk V, Tang Y, et al.
for Duchenne muscular dystrophy. Regen Med 2007; 2: 275-88. Transforming growth factorbeta1 induces the differentiation of
[71] Davies KE, Ground MD. Treating muscular dystrophy with stem myogenic cells into fibrotic cells in injured skeletal muscle: a key
cells? Cell 2007; 127: 1304-6. event in muscle fibrogenesis. Am J Pathol 2004; 164: 1007-19.
[72] Bretag A. Too much hype, not enough hope: Are balanced [94] Olson EN, Sternberg E, Hu JS, Spizz G, Wilcox C, et al.
reporting and proper controls too much to expect from therapeutic Regulation of myogenic differentiation by type  transforming
studies in animal models of neuromuscular diseases that presage growth factor. J Cell Biol 1986; 103: 1799-805.
clinical trials in humans? Neuromuscul Disord 2007; 17: 203-5. [95] Bernasconi P, Torchiana E, Confalonieri P, Brugnoni R, Barresi R,
Mora M, et al. Expression of transforming growth factor-beta1 in
Muscular Dystrophies Current Pharmaceutical Design, 2010, Vol. 16, No. 8 987

dystrophic patient muscles correlates with fibrosis. J Clin Invest -induced failure of muscle regeneration in multiple myopathic
1995; 96: 1137-44. states. Nat Med 2007; 13: 204-10.
[96] Chen YW, Nagaraju K, Bakay M, McIntyre O, Rawat R, Shi R, [99] Florentin M, Liberopoulos EN, Mikhailidis DP, Elisaf MS. Fibrate-
et al. Early onset of inflammation and later involvement of TGF associated adverse effects beyond muscle and liver toxicity. Curr
in Duchenne muscular dystrophy. Neurology 2005; 65: 826-34. Pharm Des 2008; 14(6): 574-87.
[97] Zanotti S, Negri T, Cappelletti C, Bernasconi P, Canioni E, Di [100] Fu DX, Foss CA, Nimmagadda S, Ambinder RF, Pomper MG.
Blasi C, et al. Decorin and biglycan expression is differentially Imaging virus-associated cancer. Curr Pharm Des 2008; 14(28):
altered in several muscular dystrophies. Brain 2005; 128: 2546-55. 3048-65.
[98] Cohn RD, van Erp C, Habashi JP, Soleimani AA, Klein EC, Lisi [101] Rovere-Querini P, Brunelli S, Clementi E, Manfredi AA. Cell
MT, et al. Angiotensin II type 1receptor blockade attenuates TGF- death: tipping the balance of autoimmunity and tissue repair. Curr
Pharm Des 2008; 14(3): 269-77.

Received: November 17, 2009 Accepted: December 4, 2009

You might also like