+ All Categories
Home > Documents > Exp Physiol 2011 Fairclough Expphysiol.2010.053025

Exp Physiol 2011 Fairclough Expphysiol.2010.053025

Date post: 02-Jun-2018
Category:
Upload: carlos-fruehbeck-moreno
View: 224 times
Download: 0 times
Share this document with a friend

of 31

Transcript
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    1/31

    Progress in therapy for Duchenne muscular dystrophy

    Rebecca J. Fairclough*, Akshay Bareja*, Kay E. Davies#

    MRC Functional Genomics Unit, Department of Physiology Anatomy and Genetics, Universityof Oxford OX1 3PT, UK

    *equal contribution

    # corresponding author

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    2/31

    Abstract

    Duchenne muscular dystrophy is a devastating muscular dystrophy of childhood.

    Mutations in the dystrophin gene destroy the link between the internal muscle filaments

    and the extracellular matrix resulting in severe muscle weakness and progressive muscle

    wasting. There is currently no cure, and, whilst palliative treatment has improved, affected

    boys are normally in a wheelchair by twelve years of age and die from respiratory or

    cardiac complications in their 20s-30s. Therapies currently being developed include

    mutation-specific treatments, DNA- and cell-based therapies, and drugs which aim to

    modulate cellular pathways or gene expression. This review aims to provide an overview

    of the different therapeutic approaches aimed at reconstructing the dystrophinassociated

    protein complex, including restoration of dystrophin expression and upregulation of the

    functional homologue utrophin.

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    3/31

    Introduction

    Duchenne muscular dystrophy (DMD) is an X-linked genetic disease that afflicts one in

    every 3,500 males (Emery, 1992). It is caused by the absence of the protein

    dystrophin(Koenig et al., 1987). DMD patients are generally boys who exhibit early

    symptoms (around 3-5 years of age) of abnormal gait, calf muscle pseudohypertrophy

    (Emery, 1992) and the characteristic Gowers sign (weakness of the proximal muscles,

    particularly those of the lower limb; Gowers, 1985). Respiratory complications, including

    sleep-disordered breathing and apnoea, arise during the teenage years resulting in

    headaches, nausea, fatigue and poor appetite (Emery, 1992; Cox & Kunkel, 1997). Patients

    are usually wheelchair-bound by 12 years of age (Emery, 1992). Dilated cardiomyopathy

    of varying severity is evident from abnormal ECGs in most patients by 18 years of age, and

    is currently responsible for the death of approximately 10-40% of all DMD patients(Baxter, 2006). The development of scoliosis is coincident with permanent wheelchair use,

    rapidly progressing during puberty and adversely affecting respiratory function (Emery &

    Muntoni, 2003). Many patients die of respiratory failure in their twenties (Emery &

    Muntoni, 2003). Becker muscular dystrophy (BMD) is a milder disease caused by the

    reduced expression of dystrophin or expression of a partially functional protein. Some

    BMD patients only present with a cardiomyopathy and remain ambulant into their fifties or

    sixties (Yazaki et al., 1999). There is currently no cure for DMD. Prednisolone has been

    shown to increase the ambulatory period for some patients (Connolly et al., 2002) but more

    effective interventions are desperately needed.

    Dystrophin and the Dystrophin Associated Protein Complex

    Dystrophin is a 427-kDa protein that is comprised of four domains: the N-terminal domain

    (which binds actin), the rod domain (which consists of 26 spectrin-like repeats), a cysteine-

    rich domain and a C-terminal domain (Koenig et al., 1988).

    Skeletal muscle is made up of thousands of muscle fibres (myofibres), each of which

    is surrounded by a plasma membrane (sarcolemma) and an overlying basal lamina.

    Dystrophin is expressed in skeletal muscle at the sarcolemma and is enriched at the

    myotendinous junction (MTJ; the junction of muscle fibres and tendons) and the

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    4/31

    neuromuscular junction (NMJ; the junction of a motor neuron with a muscle fibre)

    (Khurana et al., 1991). Dystrophin binds to filamentous (F)-actin (a constituent member of

    the cytoskeleton) via its N-terminus and a cluster of basic repeats 11-17 in its rod domain

    (Amann et al., 1999). The protein binds dystroglycan via its cysteine-rich domain, and

    -

    dystrobrevin and

    -syntrophin via its C-terminal domain (Junget al., 1995; Yanget al.,

    1995; Sadoulet-Puccioet al., 1997). Figure 1 (taken from Davies & Nowak, 2006)

    illustrates the mechanical link dystrophin creates between the internal cytoskeleton and the

    external extracellular matrix.

    The main function of dystrophin is to stabilize the sarcolemma. Due to the elasticity

    and flexibility of its large central rod domain consisting of triple helical repeats, dystrophin

    has been proposed to protect the myofibre from contraction-induced stress (Petrof et al.,

    1993). It has also been shown to form a mechanically strong link between the sarcolemma

    and the internal cytoskeleton via its interaction with gamma-actin filaments (Rybakova et

    al., 2000). It has also been postulated to play a role in signalling pathways, and this is

    discussed further below.

    Preclinical animal models of DMD

    The most widely used DMD mouse model is the mdx mouse, which is a naturally occurring

    mutant that has a premature stop codon in exon 23 of its dystrophin gene due to a point

    mutation, resulting in the complete absence of full-length dystrophin expression. Muscle

    degeneration follows a characteristic progression in the mdx mouse, beginning with a sharp

    increase in myofibre necrosis and elevated serum creatine kinase levels around 3 weeks of

    age (McGeachie et al., 1993), which decreases in severity to a chronic low level of damage

    by 8 weeks of age (Spencer & Tidball, 1996). This persists until about 1 year of age

    following which there is a further reduction in the levels degeneration. Despite being the

    exact murine analogue of human DMD patients, mdx mice do not display a disease

    phenotype that is as severe as that of DMD patients. For example, they do not show any

    impairment in mobility throughout their lifetime and, with the exception of the diaphragm,

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    5/31

    their skeletal muscles do not display similar levels of fibrosis (Stedmanet al., 1991;

    Connollyet al., 2001; Rollandet al., 2006)

    Due to the relatively mild phenotype of the mdxmouse, many attempts have been made to

    exacerbate this phenotype by generating double knockouts. These have been reviewed in detail

    elsewhere (Willmann et al., 2009). The double knockout that has been used most extensively in

    preclinical studies for DMD is the utrophin/dystrophin double knockout (dKO, mdx; utrn-/-

    )

    which was generated by crossing mdxmice with a knock-out of the functional dystrophin

    homologue, utrophin (Deconinck et al., 1997; Grady et al., 1997; the role of utrophin is described

    in detail below). The dKO mouse displays a disease phenotype far more similar in its severity

    to that of DMD patients. Unlike mdxmice, dKO mice have a significantly reduced lifespan of 4-

    20 weeks, and display many of the hallmark symptoms of DMD, such as severe muscle

    weakness, joint contractures, kyphosis (curvature of the upper spine) and pronounced growth

    retardation post-weaning. The accurate recapitulation of the dystrophic progression of DMD in

    this mouse model, coupled with the absence of the compensatory power of utrophin, means that

    the dKO mouse can serve as a better model than the mdxmouse in which to test the effectiveness

    of therapeutic strategies. For example, this model has recently been used to test the effectiveness

    of an exon-skipping strategy (Goyenvalle et al., 2010).

    The best characterized and most widely used canine model of DMD is the Golden

    retriever (GRMD) dog model. GRMD lacks dystrophin due to incorrect splicing resulting in a

    truncated transcript (Sharp et al., 1989). These dogs display a phenotype that is very similar to

    human DMD patients. The GRMD dog has been successfully used to test the therapeutic

    potential of mesoangioblast administration (Sampaolesi et al., 2006). The major problem with

    the use of these dogs is that they display a high degree of phenotypic variation (Ambrosio et

    al., 2008).

    Pathophysiological consequences of dystrophin absence

    Since the DAPC plays many roles in the proper functioning of the skeletal muscle, the

    absence of dystrophin, and hence the impaired recruitment of the members of the DAPC to

    the sarcolemma, creates a pathophysiological cascade with numerous adverse downstream

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    6/31

    effects. Since the primary function of dystrophin is to stabilize the sarcolemma, its absence

    renders the sarcolemma vulnerable to damage-inducing factors (Petrof et al., 1993). As the

    affected organism grows in size, increased amounts of stress will be imposed on the

    sarcolemma as a result of increased load and force generation. The resulting membrane

    instability has many deleterious consequences. Transient tears in the membrane promote

    the unregulated influx of Ca2+

    ions into the sarcoplasm which is thought to have toxic

    effects primarily via its effect on mitochondrial genes (Chen et al., 2000). The continuous

    degeneration and regeneration of muscle tissue as a result of stress factors has been

    suggested to be responsible for elevated levels of fibrosis as exemplified by excess collagen

    deposition (Morrison et al., 2000). The lack of dystrophin and, hence,

    -syntrophin, results

    in the down regulation and relocalization of nNOS at the sarcolemma (Chang et al., 1996).

    nNOS activity is thought to counter the vasoconstrictive response of the muscle,

    particularly during periods of exercise (Thomaset al., 1998; Laiet al., 2009a). The

    dysregulation of blood flow as a result of its absence is therefore thought to result in

    functional ischaemia (Sander et al., 2000). One study suggests that the absence of

    dystrophin results in the downregulation of Akt, which normally functions to inhibit

    MuRF1 and MAFbx: E3 ubiquitin ligases that are involved in muscle atrophy by targeting

    components of the myofibre for proteasomal degradation (Acharyya et al., 2005). This

    study also showed that loss of dystrophin expression led to aberrant glycosylation of

    -

    dystroglycan and

    -sarcoglycan, both of which are key members of the DAPC.

    Therapies for DMD

    Numerous different therapeutic strategies have been devised to correct the various

    deleterious consequences of dystrophin absence. This review covers some of the most

    promising approaches to therapy which tackle the primary cause. Treatments which

    alleviate the secondary effects of the disease have been reviewed elsewhere (Zhou & Lu;Bogdanovichet al., 2004; Bushbyet al., 2009; Zhou & Lu, 2010). These strategies can be

    divided into four categories: cell therapy (myoblast transplantation and stem cell therapy),

    gene replacement therapy, mutation-specific approaches (exon skipping and suppression of

    premature termination codons) and utrophin upregulation.

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    7/31

    Cell therapy

    Cell therapy involves the delivery of cells that make new muscle to diseased areas. These

    can either be muscle precursor cells or stem cells that have the ability to differentiate into

    muscle cells. The cell therapy strategy that has experienced the most success so far has

    been the transplantation of myoblasts into diseased tissue.

    Myoblasts are muscle precursor cells that can differentiate to form myofibres.

    Myoblast transplantation (MT) involves the derivation of myoblast cells from healthy

    donor skeletal muscle, expansion of these cells in culture, and administration to dystrophic

    tissue. The incorporation of donor myoblasts into recipient myofibres results in a

    phenomenon known as gene complementation, which means the expression of both

    exogenous and host genes in the myofibre syncitium (Watt et al., 1982). Following good

    transplantation results in immunosuppressed mdxmice (Partridgeet al., 1989; Morgan &

    Partridge, 1992; Vilquinet al., 1994) great expectation saw this therapy propelled into

    human trials. Sadly, these trials yielded only limited positive results, with a maximum of

    10 % donor dystrophin-positive fibres in one out of twelve patients (Mendell et al., 1995).

    This poor result has been attributed to poor immunosuppression combined with insufficient

    numbers of transplanted cells and insufficient distribution of cells (Gussoniet al., 1992;

    Karpatiet al., 1993; Tremblayet al., 1993a; Tremblayet al., 1993b; Mendellet al., 1995).

    Since then improvements in tissue repopulation were achieved in DMD animal models

    through the use of muscle slice grafts (Fan et al., 1996), although translation into clinical

    trials would of course be limited to larger more surgically accessible dystrophic muscles. It

    has become clear that the success of MT transplantation is also closely related to the choice

    of immunosuppressive drug used (Hong et al., 2002). Current hopes are pinned on the use

    of donor genetically modified myoblasts to reduce the patient immune response (Li et al.,

    2005; Benchaouiret al., 2007; Kazukiet al., 2010). Lots of studies have been performed to

    identify factors that may improve myoblast migration through adult tissues (Fakhfakhetal.; Gerardet al.; Smythe & Grounds, 2000; Smytheet al., 2001; Smythe & Grounds,

    2001). Other studies suggest increasing the number of cells transplanted may not be

    beneficial (Pellegrini & Beilharz); moreover, a recent study by Hall et al. (Hall et al.)

    suggested that only a very small number of satellite cells are able to repopulate muscle

    successfully. This study was performed using a small number of satellite cells that were

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    8/31

    cultured in vitro and subsequently transplanted into muscle whilst still attached to the

    myofibres. Perhaps the microenvironment of the myofibre provides novel elements that are

    crucial for the subsequent survival and proliferation of the satellite cells following

    transplantation?

    It was initially thought that only satellite cells were responsible for the growth and

    maintenance of skeletal muscle. There are, however, other cells found in skeletal muscle

    that have myogenic potential, namely muscle-derived stem cells (MDSCs), muscle side-

    population (mSP) cells and muscle-derived CD133+ progenitors. Certain stem cells derived

    from non-muscle have also been shown to be able to participate in myogenesis. For

    example, bone marrow-derived mesenchymal stem cells (MSCs) can differentiate into

    mesodermal cells, including myoblasts (Bhagavati & Xu, 2004). MSCs have the

    advantages of being able to fuse with and genetically complement dystrophic muscle,

    possessing anti-inflammatory properties, and producing factors that enhance the activity of

    endogenous repair cells (Ichim et al., 2010). However, most of the studies using MSCs in

    animal models did not show an appreciable improvement in muscle contractile force (Ichim

    et al., 2010). Another type of stem cell whose use has been proposed to be a possible

    therapeutic option is the mesoangioblast cell. This is a multipotent progenitor of

    mesodermal tissue whose transplantation in dystrophic dogs has been shown to result in the

    re-expression of dystrophin in myofibres and an improvement in muscle contractile force

    (Sampaolesi et al., 2006).

    A major limitation that needs to be overcome if cell therapy is going to be used to

    successfully treat DMD patients is the inability of intravenous and intraperitoneal injections

    of myoblasts to repopulate dystrophic muscle (Partridge, 1991), necessitating the use of

    intramuscular injections. The major disadvantage of this delivery method is that the

    myoblasts fuse mainly with the myofibres that neighbor the site of injection, resulting in

    restricted zones of muscle growth and recovery (El Fahimeet al., 2000; Torrenteet al.,

    2000; El Fahimeet al., 2002; Skuket al., 2002). Of course, local transfer also makes

    treatment of less accessible muscles, such as the diaphragm, virtually impossible.

    Stem cell therapy in clinical trial for DMD includes CD133+ (Torrente et al., 2007)

    and mesoangioblasts (in progress; under the direction of Giulio Cossu).

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    9/31

    Conventional gene replacement strategies

    Over recent years there has been significant advancement in research towards an effective

    therapy for DMD using direct gene replacement approaches. This strategy is limited by the

    large size (14kb) of the dystrophin gene. However, there are very mildly affected BMD

    patients who are ambulant into their 50s and 60s who lack up to 50% of the gene sequence.

    Significantly, these deletions occur in the rod domain of the protein encoding spectrin

    repeats (Englandet al., 1990; Loveet al., 1991; Matsumuraet al., 1994). These gene

    deletions have formed the basis of the generation of mini- and micro-dystrophin genes with

    increasingly large sections of the rod domain removed to enable them to be cloned into

    AAV vectors (for a detailed review see (Goyenvalle & Davies, 2011a)). Systemic delivery

    of mini-dystrophin genes with AAV9 has been very effective in mouse and dog (Pichavant

    et al., 2010). However, a recent clinical trial using intramuscular injections of AAV2 into

    DMD patients did not result in good restoration of dystrophin expression and highlighted

    the potential for T cell immunity to dystrophin epitopes (Mendellet al., 2010). An

    alternative strategy is the delivery of micro- genes encoding the dystrophin-related protein

    utrophin (see below) which should not elicit an immune response.

    Problems faced by this type of approach to therapy include the production of

    sufficient quantities of virus and circumventing the immune response encountered through

    the introduction of a previously absent protein. Additionally, these shortened genes might

    not be capable of completely rescuing the DAPC defect because their protein products will

    be unlikely to recapitulate all of dystrophins interactions with members of the DAPC. For

    example, full length dystrophin (together with syntrophin) recruits neuronal nitric oxide

    synthase (nNOS) to the sarcolemma (Hillieret al., 1999; Kameyaet al., 1999; Tochioet

    al., 1999; Adamset al., 2000; Laiet al., 2009a). nNOS localized at the membrane

    counteracts

    -adrenergic vasoconstriction during muscle contraction by allowing diffusion

    of nitric oxide. Failure to do this aggravates muscle disease by resulting in functional

    ischaemia (Sander et al., 2000). It has been shown that the sarcolemmal targeting of nNOS

    is dependent on the spectrin-like repeats 16 and 17 of the rod domain (Lai et al., 2009b),

    which means that any truncated dystrophin transgene that lacks this region is not likely to

    be an entirely functional replacement.

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    10/31

    A phase I safety study of mini-dystrophin is currently being performed under the

    direction of Asklepios Biopharmaceutical Inc. and Jerry Mendell.

    Suppression of premature termination codons

    Studies have shown that mammalian cells treated with high concentrations of

    aminoglycoside antibiotics like gentamicin promote readthrough of premature stop codons

    by interfering with the ribosomes ability to recognize these stop signals (Manuvakhova et

    al., 2000). Gentamicin treatment could therefore be applied to DMD patients with nonsense

    mutations in their dystrophin gene. A six-month trial of gentamicin on DMD patients

    resulted in a significant increase in dystrophin expression in skeletal muscle (with the

    highest level approaching 13-15% of normal expression in one individual) and decreased

    levels of creatine kinase - a marker of muscle damage. However, one of the individuals

    developed an immune response to the newly-expressed dystrophin protein (Malik et al.,

    2010). Additional problems with gentamicin include its lack of potency and possible toxic

    effects (Welch et al., 2007).

    PTC124 is a synthetic drug that has been shown to have the ability to induce

    readthrough of premature but not normal stop codons. Treatment of mdx mice with

    PTC124 resulted in a significant increase in dystrophin expression in skeletal muscle and

    partial (yet significant) recovery of muscle contractile force (Welch et al., 2007). Some of

    the results from this study have however been recently called into question and a Phase IIb

    clinical trial of PTC124 has recently been suspended (Finkel, 2010; Guglieri and Bushby,

    2010). It is estimated that 13 % of DMD patients have nonsense mutations and could

    therefore be treated by nonsense suppression.

    Exon skipping

    Exon skipping is a promising RNA based approach which could target up to 83% of DMD

    patients with deletions (Aartsma-Ruset al., 2009). It relies on synthesised antisense

    oligonucleotides (AONs) carried into the cell using a range of different chemical

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    11/31

    backbones. These include peptide nucleic acids (PNA), 2-O-methyl-phosphorothiate-

    AONs (2OMeAO), phosphorodiamidate morpholino oligomer (PMO) and cell penetrating

    peptide-conjugated PMO (PPMO). AOs alter splicing by either sterically blocking splice

    enhancer sequences or by altering secondary mRNA structure folding. Success with this

    technique has been achieved in cell culture, and trials in mdx (Alter et al., 2006) and dKO

    (Goyenvalle et al., 2010) have ameliorated the disease phenotypes of both mouse models.

    Exon 51 skipping represents the therapy with the potential to target the largest percentage

    (13 %) of DMD patients and, for this reason, clinical trials are currently concentrating on

    this target. A Phase III systemic trial of GSK2402968 (GSK; originally developed by

    Prosensa, Netherlands and called PRO51), based on the 2OMeAO chemistry and targeting

    exon 51, is ongoing. A phase I/II clinical trial of AVI-4658 (AVI Biopharma, USA), again

    targeting exon 51 and based on the PMO backbone, has recently been completed. Both

    have demonstrated good tolerability of the drug and restoration of dystrophin expression at

    the sarcolemma (Goemanset al., 2010; Shrewsburyet al., 2010). However, poor cellular

    uptake, relative rapid clearance from the circulation, variable skipping efficiency between

    muscles and poor efficacy in the heart are just several of the challenges standing between

    these drugs and routine clinical use (for detailed review see (Goyenvalle & Davies,

    2011a)). Furthermore, there are indications from preclinical trials that doses required for

    functional improvement may be too high to make lifelong treatment economically possible

    for the majority of patients. In view of this, much hope is being placed on the development

    of PPMOs which demonstrate enhanced internalisation by muscle cells (Moulton &

    Moulton, 2010) at much lower doses (Jearawiriyapaisarn et al., 2008; Yin et al., 2008; Yin

    et al., 2009) and demonstrate efficacy in the heart (Jearawiriyapaisarn et al., 2010; Wu et

    al., 2010; Wu et al., 2008; Yin et al., 2008b).

    A novel approach to exon skipping in DMD therapy which is being further

    developed in our laboratory by Aurelie Goyenvalle uses U7, a non spliceosome smallnuclear RNA (snRNA), normally involved in the processing of the histone mRNA 3 end,

    to enhance the delivery of antisense sequences (Goyenvalle & Davies, 2011b). By slightly

    modifying the binding site for Sm/Lsm proteins, U7 can be converted into a versatile tool

    for splicing modulation. Delivery of the appropriately modified U7 snRNA using an

    adeno-associated virus has demonstrated widespread dystrophin restoration in both the mdx

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    12/31

    mouse (Goyenvalleet al., 2004) and the GRMD (Vulin et al., 2011) models of DMD

    following only a single dose. Through vectorizing the delivery of AONs, the resulting long

    term restoration of dystrophin has obvious advantages over the need for repeated dosing

    following administration of synthetic AONs.

    Exon skipping holds a lot of promise but many hurdles remain. The fact that specific

    oligonucleotide sequences will be required to target each individual exon that needs to be

    skipped means that the regulatory authorities may consider each individual AON as a new

    drug. There will be a need to target all muscle types, including the heart, as a minimum of

    20 % dystrophin restoration has been postulated to be required for clinical efficacy

    (Chamberlain, 1997). Repeated administration of the AOs may be required because of their

    short half-life in muscle tissue, although this is not true for PMOs (for extensive review see

    (Manzur & Muntoni, 2009)). Multi-exon skipping, which involves the exclusion of

    multiple contiguous exons, has been proposed to be a way to circumvent the regulatory

    constraints of individual drugs for individual exons by allowing the administration of the

    same AO to patients with different mutational defects (Goyenvalle & Davies, 2011b).

    Pharmacological upregulation of the dystrophin related protein, utrophin

    Utrophin is an autosomal protein encoded by a gene on chromosome 6 in humans. The full-

    length protein has 3,433 amino acids with a predicted molecular mass of 395 kDa

    (compared to 3,678 amino acids and 427 kDa for dystrophin). The primary structure of

    utrophin is very similar to that of dystrophin, with the N-terminal, cysteine-rich and C-

    terminal domains displaying significant structural similarity, being 80 % identical to each

    other (Tinsley et al., 1992). Unlike dystrophin which is expressed in muscle with lower

    levels in brain, utrophin is ubiquitously expressed. Unlike dystrophin, which is expressed

    throughout the sarcolemma of adult skeletal muscle cells, utrophin is only expressed at the

    NMJ and MTJ, although it is found at the sarcolemma in early human development (Clerk

    et al., 1993; Tome et al., 1994). Utrophin is also expressed across the entire sarcolemma in

    developing and regenerating muscle (Khuranaet al., 1991; Tinsleyet al., 1992; Clerket

    al., 1993; Ponset al., 1993) and in the skeletal muscle of mdxmice and DMD patients

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    13/31

    (Mizuno et al., 1993). Importantly, a significant inverse correlation between utrophin

    expression and disease severity in DMD has been found (Kleopa et al., 2006).

    Utrophin shares many of the same binding partners as dystrophin. The C-terminus

    of utrophin has been shown to bind to members of the DAPC, such as

    -dystrobrevin-1

    (Peters et al., 1998) and

    -dystroglycan (Ishikawa-Sakurai et al., 2004). Whilst both

    utrophin and dystrophin bind

    -dystroglycan with similar affinity through their ZZ domain,

    their modes of binding slightly differ (Ishikawa-Sakurai et al., 2004). Also, like

    dystrophin, utrophin binds to cytoskeletal F-actin. However, again, as in the case of

    -

    dystroglycan binding, the mode of contact is different. Whilst dystrophin binding occurs

    through two independent actin-binding sites, utrophin binding occurs through one

    continuous stretch of the protein (Amannet al., 1998; Rybakova & Ervasti, 2005).

    Mechanistically, it has been postulated that this difference enables dystrophin to act as a

    molecular shock absorber to dampen elastic recoil during contraction or stretch. In

    contrast, utrophin may normally function as a molecular ruler to help define the length of

    costameric actin filaments during muscle development. The strong similarities between

    dystrophin and utrophin prompted the hypothesis that utrophin upregulation could lead to

    the functional replacement of dystrophin at the sarcolemma. Over expression of minigenes

    from an utrophin transgene driven by the human skeletal actin promoter in the mdxmouse

    has been shown to prevent the dystrophic pathology (Tinsleyet al., 1996; Gilbertet al.,

    1999). This model was named the Fiona mouse and is a transgenic mdxmouse that

    expresses full-length utrophin in skeletal muscle at a 3-4 fold higher level than in non-

    transgenic mdxmouse muscle. These mice are phenotypically indistinguishable from wild-

    type mice in sedentary conditions and show complete correction of the dystrophic

    phenotype, displaying even myofibre size, normal levels of muscle degeneration and

    regeneration, and similar levels of force production by the extensor digitorum longus

    (EDL) muscle compared to wild-type muscle (Tinsley et al., 1998). Whilst Li et al. (Li etal., 2010) recently demonstrated that utrophin does not completely compensate in exercised

    mice, critically, it does stabilise the sarcolemma (Bareja, 2011). Delivery of utrophin using

    intramuscular injections of adenoviruses in the GRMD also showed an improvement of

    phenotype (Cerletti et al., 2003). In the mdxmouse utrophin expression initiated earlier in

    development has a more potent therapeutic effect than when initiated later, exactly as is

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    14/31

    observed for dystrophin (Liu et al., 2005; Squire et al., 2002). Crucially for drug

    development, body-wide overexpression of utrophin at levels which prevent the pathology

    does not have detrimental effects in the mdxmouse (Fisher et al., 2001). These studies

    collectively indicate that upregulating utrophin could have a powerful therapeutic effect in

    DMD patients.

    Utrophin expression is predominantly driven by two promoters: A and B (Burton et

    al., 1999). The utrophin A promoter is responsible for the skeletal muscle-specific

    expression of utrophin and promoter B drives expression in endothelial cells (Weir et al.,

    2002). The core promoter A element contains recognition sites for the transcription factors

    Ap2, Sp1 and Sp3, which orchestrate basal transcriptional activity (Perkins et al., 2001).

    Importantly, the upstream promoter consists of motifs including the N-box, E-box and

    NFAT site, which have all been shown to influence the expression of utrophin (Angus et

    al., 2005; Gramolini et al., 1999; Perkins et al., 2001). The N-box localises the expression

    of utrophin to the NMJ (Dennis et al., 1996). More recently, a PPRE element has been

    described which is known to be important in regulating the transcription of genes involved

    in lipid metabolism, oxidative respiration and promoting the formation of slower, more

    oxidative muscle fibers (Miura et al., 2009).

    A range of strategies have been used to upregulate utrophin. These include the use

    of heregulin, which acts via the N-box motif of the utrophin A promoter (Krag et al., 2004)

    and L-arginine, which results in an increase in utrophin expression as a result of increased

    production of nNOS (Voisin & de la Porte, 2005). Jazz is an artificially generated

    transcription factor designed to target a specific region in the utrophin A promoter (Corbi et

    al., 2000; Di Certo et al., 2010), administration of a PPAR/

    agonist GW501516 results

    in a fast-to-slow fibre-type switch and concomitant increase in utrophin expression in mdx

    mice (Miura et al., 2009). Biglycan is an extracellular matrix protein that most likely

    increases utrophin expression by recruiting the protein to the sarcolemma without having

    an effect on transcript levels (Amenta et al., 2011). Overexpression of CT-GalNac

    transferase in mdxmice skeletal muscle results in increased carbohydrate glycosylation at

    the sarcolemma and a resultant increase in extrasynaptic utrophin expression (Durko et al.,

    2010). TAT-utrophin, a recombinant utrophin protein modified with the HIV-derived TAT

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    15/31

    protein transduction domain, improves delivery across the cell membrane (Sonnemann et

    al., 2009). Overexpression of RhoA, a small GTPase, results in an increase in utrophin

    expression with no change in transcription levels (Gauthier-Rouviere & Bonet-Kerrache,

    2009). Although all these experiments have yielded positive results, none have been able to

    achieve complete amelioration of the mdxdystrophic phenotype and the 3-4 fold

    upregulation of utrophin thought to be necessary for complete correction.

    Our laboratory has used the approach of finding drugs which increase utrophin

    levels by exploiting a stable H2K mdxmyoblast cell line (Morgan et al., 1994) which

    encodes 8.4 kb of the mouse utrophin promoter A lying 5' of the transcription start site and

    linked to a luciferase reporter gene (Davies, Russell and Davies unpublished; Khurana &

    Davies, 2003; Tinsleyet al., 2011). An initial screen of 5000 small compounds, carefully

    designed to cover a broad range of biological space and omiting any structures containing

    reactive or cytotoxic functionalities, was able to identify candidates which upregulated

    utrophin gene expression in a dose-dependent manner. A secondary assay was used to

    confirm that active compounds increased utrophin mRNA (and were not acting through

    stablisation of the luciferase reporter). A final in vitroassay using human DMD patient

    cell lines then confirmed increased utrophin protein expression. The in vivoeffect of

    positive compounds was then assessed following intraperitoneal administration or oral

    gavage in the mdxmouse. This work was done in partnership with Summit plc, who used

    the cell line to find drugs which increased utrophin levels 5-fold. The chemistry was

    optimized and resulted in one drug, SMT C1100, which showed therapeutic potential as

    assessed by its prevention of pathology in the mdxmouse (Tinsley et al., 2011).

    SMT C1100 targets the primary cause of the disease by reducing the level of

    muscle membrane damage, as demonstrated by a reduction in force drop following

    eccentric contractions (Tinsleyet al., 2011). Serum creatine kinase, muscle fibrosis and

    necrosis are also reduced indicating that SMT C1100 diminishes the catastrophic secondary

    pathology associated with the disease. Even when efficacy was tested in the more severe

    exercised mdxmouse model, SMT C1100 treatment afforded increased muscle strength, a

    partial reduction in the resistance to fatigue, and a significant amelioration of calcium-

    dependent functional parameters (Tinsleyet al., 2011). Interestingly, the drug also

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    16/31

    demonstrated a synergistic effect when administered with prednisolone (Tinsleyet al.,

    2011) the current gold standard in treatment of DMD patients in the clinic. SMT C1100

    was taken into Phase I trials by BioMarin pharmaceuticals (as BMN-195). Although there

    were no safety issues, the plasma levels of the drug were not high enough for the trials to

    continue into patients. New formulations of the drug are currently being explored by

    Summit plc with a view to taking this compound back into the clinic.

    SMT C1100 demonstrates the principle of finding drugs which upregulate utrophin

    and more screens are ongoing. Future screens will be based on a cell line which has the

    luciferase gene knocked into exon 7 of the endogenous gene (Fairclough and Davies,

    unpublished). This will enable screening of the promoter in its genomic context and

    therefore should include most, if not all, of the regulatory elements, therefore better

    mimicking the in vivosituation.

    Concluding remarks

    It is more than 20 years since the identification of the dystrophin gene and the development

    of therapy has taken a long time. In spite of the challenges that remain, the current clinical

    trials are looking very promising. Even if one strategy does not work efficiently enough, it

    may be possible to combine these approaches. The next phase of DMD research looks to

    be a very exciting one for scientists, clinicians and patients.

    Acknowledgements

    We are grateful to the Medical Research Council, Muscular Dystrophy Campaign,

    Muscular Dystrophy Association USA, Association Francaise contre les Myopathies and

    the Clarendon fund for their support of our work. We would also like to thank Aurelie

    Goyenvalle for her comments on the manuscript and helpful suggestions towards the

    figures.

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    17/31

    References

    Aartsma-Rus A, Fokkema I, Verschuuren J, Ginjaar I, van Deutekom J, van Ommen GJ & denDunnen JT (2009). Theoretic applicability of antisense-mediated exon skipping for

    Duchenne muscular dystrophy mutations.Hum Mutat30, 293-299.

    Acharyya S, Butchbach MER, Sahenk Z, Wang HT, Saji M, Carathers M, Ringel MD,

    Skipworth RJE, Fearon KCH, Hollingsworth MA, Muscarella P, Burghes AHM, Rafael-

    Fortney JA & Guttridge DC (2005). Dystrophin glycoprotein complex dysfunction: Aregulatory link between muscular dystrophy and cancer cachexia. Cancer Cell8, 421-

    432.

    Adams ME, Kramarcy N, Krall SP, Rossi SG, Rotundo RL, Sealock R & Froehner SC (2000).

    Absence of alpha-syntrophin leads to structurally aberrant neuromuscular synapsesdeficient in utrophin.J Cell Biol150, 1385-1398.

    Alter J, Lou F, Rabinowitz A, Yin HF, Rosenfeld J, Wilton SD, Partridge TA & Lu QL (2006).Systemic delivery of morpholino oligonucleotide restores dystrophin expression

    bodywide and improves dystrophic pathology.Nature Medicine12, 175-177.

    Amann KJ, Guo AW & Ervasti JM (1999). Utrophin lacks the rod domain actin binding activity

    of dystrophin.J Biol Chem274, 35375-35380.

    Amann KJ, Renley BA & Ervasti JM (1998). A cluster of basic repeats in the dystrophin rod

    domain binds F-actin through an electrostatic interaction.Journal of Biological

    Chemistry273, 28419-28423.

    Ambrosio CE, Valadares MC, Zucconi E, Cabral R, Pearson PL, Gaiad TP, Canovas M, VainzofM, Miglino MA & Zatz M (2008). Ringo, a Golden Retriever Muscular Dystrophy(GRMD) dog with absent dystrophin but normal strength.Neuromuscul Disord18, 892-

    893.

    Amenta AR, Yilmaz A, Bogdanovich S, McKechnie BA, Abedi M, Khurana TS & Fallon JR

    (2011). Biglycan recruits utrophin to the sarcolemma and counters dystrophic pathology

    in mdxmice. Proc Natl Acad Sci U S A108, 762-767.

    Bareja A (2011). Utrophin upregulation and microRNAs - two avenues of Duchenne muscular

    dystrophy therapy research

    InDepartment of Physiology, Anatomy and Genetics, vol. D.Phil. Oxford University, Oxford.

    Baxter P (2006). Treatment of the heart in Duchenne muscular dystrophy. Dev Med Child Neurol48, 163.

    Benchaouir R, Meregalli M, Farini A, D'Antona G, Belicchi M, Goyenvalle A, Battistelli M,Bresolin N, Bottinelli R, Garcia L & Torrente Y (2007). Restoration of human dystrophin

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    18/31

    following transplantation of exon-skipping-engineered DMD patient stem cells into

    dystrophic mice. Cell Stem Cell1, 646-657.

    Bhagavati S & Xu W (2004). Isolation and enrichment of skeletal muscle progenitor cells from

    mouse bone marrow.Biochem Biophys Res Commun318, 119-124.

    Bogdanovich S, Perkins KJ, Krag TO & Khurana TS (2004). Therapeutics for Duchenne

    muscular dystrophy: current approaches and future directions.J Mol Med82, 102-115.

    Burton EA, Tinsley JM, Holzfeind PJ, Rodrigues NR & Davies KE (1999). A second promoter

    provides an alternative target for therapeutic up-regulation of utrophin in Duchennemuscular dystrophy. Proc Natl Acad Sci U S A96, 14025-14030.

    Bushby K, Lochmuller H, Lynn S & Straub V (2009). Interventions for muscular dystrophy:molecular medicines entering the clinic.Lancet374, 1849-1856.

    Cerletti M, Negri T, Cozzi F, Colpo R, Andreetta F, Croci D, Davies KE, Cornelio F, Pozza O,Karpati G, Gilbert R & Mora M (2003). Dystrophic phenotype of canine X-linked

    muscular dystrophy is mitigated by adenovirus-mediated utrophin gene transfer. Gene

    Ther10, 750-757.

    Chamberlain JS (1997). Dystrophin levels required for genetic correction of Duchenne muscular

    dystrophy.Basic and Applied Myology7, 257-255.

    Chang WJ, Iannaccone ST, Lau KS, Masters BS, McCabe TJ, McMillan K, Padre RC, Spencer

    MJ, Tidball JG & Stull JT (1996). Neuronal nitric oxide synthase and dystrophin-deficient muscular dystrophy. Proc Natl Acad Sci U S A93, 9142-9147.

    Chen YW, Zhao P, Borup R & Hoffman EP (2000). Expression profiling in the muscular

    dystrophies: identification of novel aspects of molecular pathophysiology.J Cell Biol

    151, 1321-1336.

    Clerk A, Morris GE, Dubowitz V, Davies KE & Sewry CA (1993). Dystrophin-related protein,

    utrophin, in normal and dystrophic human fetal skeletal muscle.Histochem J25, 554-561.

    Connolly AM, Keeling RM, Mehta S, Pestronk A & Sanes JR (2001). Three mouse models ofmuscular dystrophy: the natural history of strength and fatigue in dystrophin-,

    dystrophin/utrophin-, and laminin alpha2-deficient mice.Neuromuscul Disord11, 703-

    712.

    Connolly AM, Schierbecker J, Renna R & Florence J (2002). High dose weekly oral prednisone

    improves strength in boys with Duchenne muscular dystrophy.Neuromuscul Disord12,917-925.

    Cox GF & Kunkel LM (1997). Dystrophies and heart disease. Curr Opin Cardiol12, 329-343.

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    19/31

    Davies KE & Nowak KJ (2006). Molecular mechanisms of muscular dystrophies: old and newplayers.Nat Rev Mol Cell Biol7, 762-773.

    Deconinck AE, Rafael JA, Skinner JA, Brown SC, Potter AC, Metzinger L, Watt DJ, Dickson

    JG, Tinsley JM & Davies KE (1997). Utrophin-dystrophin-deficient mice as a model forDuchenne muscular dystrophy. Cell90, 717-727.

    Dennis CL, Tinsley JM, Deconinck AE & Davies KE (1996). Molecular and functional analysis

    of the utrophin promoter.Nucleic Acids Res24, 1646-1652.

    Durko M, Allen C, Nalbantoglu J & Karpati G (2010). CT-GalNAc transferase overexpression in

    adult mice is associated with extrasynaptic utrophin in skeletal muscle fibres.J Muscle

    Res Cell Motil31, 181-193.

    El Fahime E, Mills P, Lafreniere JF, Torrente Y & Tremblay JP (2002). The urokinase

    plasminogen activator: an interesting way to improve myoblast migration following theirtransplantation.Exp Cell Res280, 169-178.

    El Fahime E, Torrente Y, Caron NJ, Bresolin MD & Tremblay JP (2000).In vivomigration oftransplanted myoblasts requires matrix metalloproteinase activity.Exp Cell Res258, 279-

    287.

    Emery AEH (1992).Duchenne Muscular Dystrophy, vol. 24. Oxford University Press, Oxford.

    Emery AEH & Muntoni F (2003).Duchenne Muscular Dystrophy. Oxford University Press,Oxford.

    Fakhfakh R, Lamarre Y, Skuk D & Tremblay JP Losartan enhances the success of myoblast

    transplantation. Cell Transplant.

    Fan Y, Beilharz MW & Grounds MD (1996). A potential alternative strategy for myoblast

    transfer therapy: the use of sliced muscle grafts. Cell Transplant5, 421-429.

    Gauthier-Rouviere C & Bonet-Kerrache A (2009). RhoA leads to up-regulation and

    relocalization of utrophin in muscle fibers.Biochem Biophys Res Commun384, 322-328.

    Gerard C, Forest MA, Beauregard G, Skuk D & Tremblay JP Fibrin gel improves the survival of

    transplanted myoblasts. Cell Transplant.

    Gilbert R, Nalbantoglu J, Petrof BJ, Ebihara S, Guibinga GH, Tinsley JM, Kamen A, Massie B,

    Davies KE & Karpati G (1999). Adenovirus-mediated utrophin gene transfer mitigates

    the dystrophic phenotype of mdxmouse muscles.Hum Gene Ther10, 1299-1310.

    Goemans NM, Tulinius M, Buyse G, Wilson R, de Kimpe R, van Deutekom JCT & Campion G

    (2010). 24 week follow-up data from a phase I/IIa extension study of

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    20/31

    PRO051/GSK2402968 in subjects with Duchenne muscular dystrophy.Neuromuscular

    Disorders20, 639.

    Gowers WR (1895).A manual of the nervous system.2nd edn, vol. 1, Philadelphia.

    Goyenvalle A, Babbs A, Powell D, Kole R, Fletcher S, Wilton SD & Davies KE (2010).Prevention of Dystrophic Pathology in Severely Affected Dystrophin/Utrophin-deficient

    Mice by Morpholino-oligomer-mediated Exon-skipping.Molecular Therapy18, 198-205.

    Goyenvalle A & Davies KE (2011a). Challenges to oligonucleotides-based therapeutics for

    Duchenne muscular dystrophy. Skeletal muscle1.

    Goyenvalle A & Davies KE (2011b). Engineering exon-skipping vectors expressing U7 snRNA

    constructs for Duchenne muscular dystrophy gene therapy.Methods in MolecularBiology 709, 179-196.

    Grady RM, Teng H, Nichol MC, Cunningham JC, Wilkinson RS & Sanes JR (1997). Skeletaland cardiac myopathies in mice lacking utrophin and dystrophin: a model for Duchenne

    muscular dystrophy. Cell90, 729-738.

    Gussoni E, Pavlath GK, Lanctot AM, Sharma KR, Miller RG, Steinman L & Blau HM (1992).

    Normal dystrophin transcripts detected in Duchenne muscular dystrophy patients after

    myoblast transplantation.Nature356, 435-438.

    Hall JK, Banks GB, Chamberlain JS & Olwin BB (2011). Prevention of muscle aging by

    myofiber-associated satellite cell transplantation. Sci Transl Med2, 57ra83.

    Hillier BJ, Christopherson KS, Prehoda KE, Bredt DS & Lim WA (1999). Unexpected modes ofPDZ domain scaffolding revealed by structure of nNOS-syntrophin complex. Science

    284, 812-815.

    Hong F, Lee J, Song JW, Lee SJ, Ahn H, Cho JJ, Ha J & Kim SS (2002). Cyclosporin A blocks

    muscle differentiation by inducing oxidative stress and inhibiting the peptidyl-prolyl-cis-

    trans isomerase activity of cyclophilin A: cyclophilin A protects myoblasts fromcyclosporin A-induced cytotoxicity. FASEB J16, 1633-1635.

    Ichim TE, Alexandrescu DT, Solano F, Lara F, Campion RD, Paris E, Woods EJ, Murphy MP,Dasanu CA, Patel AN, Marleau AM, Leal A & Riordan NH (2010). Mesenchymal stem

    cells as anti-inflammatories: Implications for treatment of Duchenne muscular dystrophy.

    Cellular Immunology260, 75-82.

    Ishikawa-Sakurai M, Yoshida M, Imamura M, Davies KE & Ozawa E (2004). ZZ domain is

    essentially required for the physiological binding of dystrophin and utrophin to beta-dystroglycan.Human Molecular Genetics13, 693-702.

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    21/31

    Jearawiriyapaisarn N, Moulton HM, Buckley B, Roberts J, Sazani P, Fucharoen S, Iversen PL &

    Kole R (2008). Sustained dystrophin expression induced by peptide-conjugatedmorpholino oligomers in the muscles of mdxmice.Mol Ther16, 1624-1629.

    Jung D, Yang B, Meyer J, Chamberlain JS & Campbell KP (1995). Identification and

    characterization of the dystrophin anchoring site on beta-dystroglycan.J Biol Chem270,27305-27310.

    Kameya S, Miyagoe Y, Nonaka I, Ikemoto T, Endo M, Hanaoka K, Nabeshima Y & Takeda S

    (1999). alpha1-syntrophin gene disruption results in the absence of neuronal-type nitric-

    oxide synthase at the sarcolemma but does not induce muscle degeneration.J Biol Chem274, 2193-2200.

    Karpati G, Ajdukovic D, Arnold D, Gledhill RB, Guttmann R, Holland P, Koch PA, ShoubridgeE, Spence D, Vanasse M & et al. (1993). Myoblast transfer in Duchenne muscular

    dystrophy.Ann Neurol34, 8-17.

    Kazuki Y, Hiratsuka M, Takiguchi M, Osaki M, Kajitani N, Hoshiya H, Hiramatsu K, Yoshino

    T, Kazuki K, Ishihara C, Takehara S, Higaki K, Nakagawa M, Takahashi K, Yamanaka S

    & Oshimura M (2010). Complete genetic correction of ips cells from Duchenne musculardystrophy.Mol Ther18, 386-393.

    Khurana TS & Davies KE (2003). Pharmacological strategies for muscular dystrophy.Nat RevDrug Discov2, 379-390.

    Khurana TS, Watkins SC, Chafey P, Chelly J, Tome FM, Fardeau M, Kaplan JC & Kunkel LM(1991). Immunolocalization and developmental expression of dystrophin related protein

    in skeletal muscle.Neuromuscul Disord1, 185-194.

    Kleopa KA, Drousiotou A, Mavrikiou E, Ormiston A & Kyriakides T (2006). Naturally

    occurring utrophin correlates with disease severity in Duchenne muscular dystrophy.

    Hum Mol Genet15, 1623-1628.

    Koenig M, Hoffman EP, Bertelson CJ, Monaco AP, Feener C & Kunkel LM (1987). Completecloning of the Duchenne muscular dystrophy (DMD) cDNA and preliminary genomic

    organization of the DMD gene in normal and affected individuals. Cell50, 509-517.

    Koenig M, Monaco AP & Kunkel LM (1988). The complete sequence of dystrophin predicts a

    rod-shaped cytoskeletal protein. Cell53, 219-228.

    Krag TO, Bogdanovich S, Jensen CJ, Fischer MD, Hansen-Schwartz J, Javazon EH, Flake AW,

    Edvinsson L & Khurana TS (2004). Heregulin ameliorates the dystrophic phenotype in

    mdxmice. Proc Natl Acad Sci U S A101, 13856-13860.

    Lai Y, Thomas GD, Yue Y, Yang HT, Li D, Long C, Judge L, Bostick B, Chamberlain JS,

    Terjung RL & Duan D (2009a). Dystrophins carrying spectrin-like repeats 16 and 17

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    22/31

    anchor nNOS to the sarcolemma and enhance exercise performance in a mouse model of

    muscular dystrophy.J Clin Invest119, 624-635.

    Lai Y, Thomas GD, Yue YP, Yang HT, Li DJ, Long C, Judge L, Bostick B, Chamberlain JS,

    Terjung RL & Duan DS (2009b). Dystrophins carrying spectrin-like repeats 16 and 17

    anchor nNOS to the sarcolemma and enhance exercise performance in a mouse model ofmuscular dystrophy.Journal of Clinical Investigation119, 624-635.

    Li D, Bareja A, Judge L, Yue Y, Lai Y, Fairclough R, Davies KE, Chamberlain JS & Duan D

    (2010). Sarcolemmal nNOS anchoring reveals a qualitative difference between

    dystrophin and utrophin.Journal of Cell Science123, 2008-2013.

    Li S, Kimura E, Fall BM, Reyes M, Angello JC, Welikson R, Hauschka SD & Chamberlain JS

    (2005). Stable transduction of myogenic cells with lentiviral vectors expressing aminidystrophin. Gene Ther12, 1099-1108.

    Malik V, Rodino-Klapac LR, Viollet L, Wall C, King W, Al-Dahhak R, Lewis S, Shilling CJ,Kota J, Serrano-Munuera C, Hayes J, Mahan JD, Campbell KJ, Banwell B, Dasouki M,

    Watts V, Sivakumar K, Bien-Willner R, Flanigan KM, Sahenk Z, Barohn RJ, Walker CM

    & Mendell JR (2010). Gentamicin-induced readthrough of stop codons in Duchennemuscular dystrophy.Ann Neurol67, 771-780.

    Manuvakhova M, Keeling K & Bedwell DM (2000). Aminoglycoside antibiotics mediatecontext-dependent suppression of termination codons in a mammalian translation system.

    Rna-a Publication of the Rna Society6, 1044-1055.

    Manzur AY & Muntoni F (2009). Diagnosis and new treatments in muscular dystrophies.

    Postgrad Med J85, 622-630.

    McGeachie JK, Grounds MD, Partridge TA & Morgan JE (1993). Age-related changes in

    replication of myogenic cells in mdxmice: quantitative autoradiographic studies.JNeurol Sci119, 169-179.

    Mendell JR, Kissel JT, Amato AA, King W, Signore L, Prior TW, Sahenk Z, Benson S,McAndrew PE, Rice R & et al. (1995). Myoblast transfer in the treatment of Duchenne's

    muscular dystrophy.N Engl J Med333, 832-838.

    Miura P, Chakkalakal JV, Boudreault L, Belanger G, Hebert RL, Renaud JM & Jasmin BJ

    (2009). Pharmacological activation of PPARbeta/delta stimulates utrophin A expression

    in skeletal muscle fibers and restores sarcolemmal integrity in mature mdxmice.HumMol Genet18, 4640-4649.

    Mizuno Y, Nonaka I, Hirai S & Ozawa E (1993). Reciprocal expression of dystrophin andutrophin in muscles of Duchenne muscular dystrophy patients, female DMD-carriers and

    control subjects.J Neurol Sci119, 43-52.

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    23/31

    Morgan JE & Partridge TA (1992). Cell transplantation and gene therapy in muscular dystrophy.

    Bioessays14, 641-645.

    Morrison J, Lu QL, Pastoret C, Partridge T & Bou-Gharios G (2000). T-cell-dependent fibrosis

    in the mdxdystrophic mouse.Laboratory Investigation80, 881-891.

    Moulton HM & Moulton JD (2010). Morpholinos and their peptide conjugates: therapeutic

    promise and challenge for Duchenne muscular dystrophy.Biochim Biophys Acta1798,2296-2303.

    Partridge TA (1991). Invited review: myoblast transfer: a possible therapy for inheritedmyopathies?Muscle Nerve14, 197-212.

    Partridge TA, Morgan JE, Coulton GR, Hoffman EP & Kunkel LM (1989). Conversion of mdxmyofibres from dystrophin-negative to -positive by injection of normal myoblasts.

    Nature337, 176-179.

    Pellegrini KL & Beilharz MW (2011). The survival of myoblasts after intramuscular

    transplantation is improved when fewer cells are injected. Transplantation91, 522-526.

    Peters MF, Sadoulet-Puccio HM, Grady RM, Kramarcy NR, Kunkel LM, Sanes JR, Sealock R &

    Froehner SC (1998). Differential membrane localization and intermolecular associations

    of alpha-dystrobrevin isoforms in skeletal muscle.Journal of Cell Biology142, 1269-1278.

    Petrof BJ, Shrager JB, Stedman HH, Kelly AM & Sweeney HL (1993). Dystrophin protects thesarcolemma from stresses developed during muscle contraction. Proc Natl Acad Sci U S

    A90, 3710-3714.

    Pons F, Nicholson LV, Robert A, Voit T & Leger JJ (1993). Dystrophin and dystrophin-related

    protein (utrophin) distribution in normal and dystrophin-deficient skeletal muscles.

    Neuromuscul Disord3, 507-514.

    Rolland JF, De Luca A, Burdi R, Andreetta F, Confalonieri P & Conte Camerino D (2006).Overactivity of exercise-sensitive cation channels and their impaired modulation by IGF-

    1 in mdxnative muscle fibers: beneficial effect of pentoxifylline.Neurobiol Dis24, 466-

    474.

    Rybakova IN & Ervasti JM (2005). Identification of spectrin-like repeats required for high

    affinity utrophin-actin interaction.Journal of Biological Chemistry280, 23018-23023.

    Rybakova IN, Patel JR & Ervasti JM (2000). The dystrophin complex forms a mechanically

    strong link between the sarcolemma and costameric actin.Journal of Cell Biology150,1209-1214.

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    24/31

    Sadoulet-Puccio HM, Rajala M & Kunkel LM (1997). Dystrobrevin and dystrophin: an

    interaction through coiled-coil motifs. Proc Natl Acad Sci U S A94, 12413-12418.

    Sampaolesi M, Blot S, D'Antona G, Granger N, Tonlorenzi R, Innocenzi A, Mognol P, Thibaud

    JL, Galvez BG, Barthelemy I, Perani L, Mantero S, Guttinger M, Pansarasa O, Rinaldi C,

    De Angelis MGC, Torrente Y, Bordignon C, Bottinelli R & Cossu G (2006).Mesoangioblast stem cells ameliorate muscle function in dystrophic dogs.Nature444,

    574-579.

    Sander M, Chavoshan B, Harris SA, Iannaccone ST, Stull JT, Thomas GD & Victor RG (2000).

    Functional muscle ischemia in neuronal nitric oxide synthase-deficient skeletal muscle ofchildren with Duchenne muscular dystrophy. Proc Natl Acad Sci U S A97, 13818-13823.

    Sharp NJ, Kornegay JN & Lane SB (1989). The muscular dystrophies. Semin Vet Med Surg(Small Anim)4, 133-140.

    Shrewsbury SB, Cirak S, Guglieri M, Bushby K & Muntoni F (2010). Current progress andpreliminary results with the systemic administration trial of AVI-4658, a novel

    phosphorodiamidate morpholino oligomer (PMO) skipping dystrophin exon 51 in

    Duchenne muscular dystrophy (DMD).Neuromuscular Disorders20, 639-640.

    Skuk D, Goulet M, Roy B & Tremblay JP (2002). Efficacy of myoblast transplantation in

    nonhuman primates following simple intramuscular cell injections: toward definingstrategies applicable to humans.Exp Neurol175, 112-126.

    Smythe GM, Davies MJ, Paulin D & Grounds MD (2001). Absence of desmin slightly prolongsmyoblast proliferation and delays fusion in vivoin regenerating grafts of skeletal muscle.

    Cell Tissue Res304, 287-294.

    Smythe GM & Grounds MD (2000). Exposure to tissue culture conditions can adversely affect

    myoblast behavior in vivoin whole muscle grafts: implications for myoblast transfertherapy. Cell Transplant9, 379-393.

    Smythe GM & Grounds MD (2001). Absence of MyoD increases donor myoblast migration intohost muscle.Exp Cell Res267, 267-274.

    Sonnemann KJ, Heun-Johnson H, Turner AJ, Baltgalvis KA, Lowe DA & Ervasti JM (2009).Functional substitution by TAT-utrophin in dystrophin-deficient mice. PLoS Med6,

    e1000083.

    Spencer MJ & Tidball JG (1996). Calpain translocation during muscle fiber necrosis and

    regeneration in dystrophin-deficient mice.Exp Cell Res226, 264-272.

    Stedman HH, Sweeney HL, Shrager JB, Maguire HC, Panettieri RA, Petrof B, Narusawa M,

    Leferovich JM, Sladky JT & Kelly AM (1991). The mdxmouse diaphragm reproduces

    the degenerative changes of Duchenne muscular dystrophy.Nature352, 536-539.

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    25/31

    Thomas GD, Sander M, Lau KS, Huang PL, Stull JT & Victor RG (1998). Impaired metabolicmodulation of alpha-adrenergic vasoconstriction in dystrophin-deficient skeletal muscle.

    Proc Natl Acad Sci U S A95, 15090-15095.

    Tinsley J, Deconinck N, Fisher R, Kahn D, Phelps S, Gillis JM & Davies K (1998). Expressionof full-length utrophin prevents muscular dystrophy in mdxmice.Nat Med4, 1441-1444.

    Tinsley JM, Blake DJ, Roche A, Fairbrother U, Riss J, Byth BC, Knight AE, Kendrick Jones J,

    Suthers GK, Love DR, Edwards YH & Davies KE (1992). Primary Structure of

    Dystrophin-Related Protein.Nature360, 591-593.

    Tinsley JM, Fairclough RJ, Storer R, Wilkes F, Potter AC, Squire SE, Powell DS, Cozzoli A,

    Capogrosso RF, Lambert A, Wilson F, Wren S, De Luca A & Davies KE (2011). Dailytreatment with SMT C1100, a novel small molecule utrophin upregulator, dramatically

    reduces the dystrophic symptoms in the mdxmouse.

    Tinsley JM, Potter AC, Phelps SR, Fisher R, Trickett JI & Davies KE (1996). Amelioration of

    the dystrophic phenotype of mdxmice using a truncated utrophin transgene.Nature384,

    349-353.

    Tochio H, Zhang Q, Mandal P, Li M & Zhang M (1999). Solution structure of the extended

    neuronal nitric oxide synthase PDZ domain complexed with an associated peptide.NatStruct Biol6, 417-421.

    Torrente Y, Belicchi M, Marchesi C, Dantona G, Cogiamanian F, Pisati F, Gavina M, GiordanoR, Tonlorenzi R, Fagiolari G, Lamperti C, Porretti L, Lopa R, Sampaolesi M, Vicentini

    L, Grimoldi N, Tiberio F, Songa V, Baratta P, Prelle A, Forzenigo L, Guglieri M,Pansarasa O, Rinaldi C, Mouly V, Butler-Browne GS, Comi GP, Biondetti P, Moggio M,

    Gaini SM, Stocchetti N, Priori A, D'Angelo MG, Turconi A, Bottinelli R, Cossu G,

    Rebulla P & Bresolin N (2007). Autologous transplantation of muscle-derived CD133+stem cells in Duchenne muscle patients. Cell Transplant16, 563-577.

    Torrente Y, El Fahime E, Caron NJ, Bresolin N & Tremblay JP (2000). Intramuscular migrationof myoblasts transplanted after muscle pretreatment with metalloproteinases. Cell

    Transplant9, 539-549.

    Tremblay JP, Bouchard JP, Malouin F, Theau D, Cottrell F, Collin H, Rouche A, Gilgenkrantz S,

    Abbadi N, Tremblay M & et al. (1993a). Myoblast transplantation between monozygotic

    twin girl carriers of Duchenne muscular dystrophy.Neuromuscul Disord3, 583-592.

    Tremblay JP, Malouin F, Roy R, Huard J, Bouchard JP, Satoh A & Richards CL (1993b).

    Results of a triple blind clinical study of myoblast transplantations withoutimmunosuppressive treatment in young boys with Duchenne muscular dystrophy. Cell

    Transplant2, 99-112.

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    26/31

    Vilquin JT, Asselin I, Guerette B, Kinoshita I, Lille S, Roy R & Tremblay JP (1994). Myoblast

    allotransplantation in mice: degree of success varies depending on the efficacy of variousimmunosuppressive treatments. Transplant Proc26, 3372-3373.

    Voisin V & de la Porte S (2005). [Pharmacological treatments for Duchenne and Becker

    dystrophies].J Soc Biol199, 17-28.

    Vulin A, Barthlmy I, Goyenvalle A, Beley C, Lorain S, Peccate C, Pitri-Rouxel F, Gentil C,Thibaud J-L, Bertoldi D, Carlier P, Dreyfus P, Danos O, Voit T, Blot S & Garcia L

    (2011). Muscle function recovery in dystrophic dog after exon skipping gene therapy.

    submitted to EMBO Molecular Medicine.

    Watt DJ, Lambert K, Morgan JE, Partridge TA & Sloper JC (1982). Incorporation of donor

    muscle precursor cells into an area of muscle regeneration in the host mouse. J Neurol Sci57, 319-331.

    Weir AP, Burton EA, Harrod G & Davies KE (2002). A- and B-utrophin have differentexpression patterns and are differentially up-regulated in mdxmuscle.J Biol Chem277,

    45285-45290.

    Welch EM, Barton ER, Zhuo J, Tomizawa Y, Friesen WJ, Trifillis P, Paushkin S, Patel M, Trotta

    CR, Hwang SW, Wilde RG, Karp G, Takasugi J, Chen GM, Jones S, Ren H, Moon YC,

    Corson D, Turpoff AA, Campbell JA, Conn MM, Khan A, Almstead NG, Hedrick J,Mollin A, Risher N, Weetall M, Yeh S, Branstrom AA, Colacino JM, Babiak J, Ju WD,

    Hirawat S, Northcutt VJ, Miller LL, Spatrick P, He F, Kawana M, Feng H, Jacobson A,

    Peltz SW & Sweeney HL (2007). PTC124 targets genetic disorders caused by nonsensemutations.Nature447, 87-U86.

    Willmann R, Possekel S, Dubach-Powell J, Meier T & Ruegg MA (2009). Mammalian animal

    models for Duchenne muscular dystrophy.Neuromuscul Disord19, 241-249.

    Yang B, Jung D, Rafael JA, Chamberlain JS & Campbell KP (1995). Identification of alpha-

    syntrophin binding to syntrophin triplet, dystrophin, and utrophin.J Biol Chem270,

    4975-4978.

    Yazaki M, Yoshida K, Nakamura A, Koyama J, Nanba T, Ohori N & Ikeda S (1999). Clinical

    characteristics of aged Becker muscular dystrophy patients with onset after 30 years.EurNeurol42, 145-149.

    Yin H, Lu Q & Wood M (2008). Effective exon skipping and restoration of dystrophinexpression by peptide nucleic acid antisense oligonucleotides in mdxmice.Mol Ther16,

    38-45.

    Yin H, Moulton HM, Betts C, Seow Y, Boutilier J, Iverson PL & Wood MJ (2009). A fusion

    peptide directs enhanced systemic dystrophin exon skipping and functional restoration in

    dystrophin-deficient mdxmice.Hum Mol Genet18, 4405-4414.

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    27/31

    Zhou L & Lu H Targeting fibrosis in Duchenne muscular dystrophy.J Neuropathol Exp Neurol69, 771-776.

    Zhou L & Lu H (2010). Targeting fibrosis in Duchenne muscular dystrophy.J Neuropathol Exp

    Neurol69, 771-776.

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    28/31

  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    29/31

  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    30/31

    Figure 1

    )by guest on September 9, 2014ep.physoc.orgDownloaded from Exp Physiol (

    http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/http://ep.physoc.org/
  • 8/11/2019 Exp Physiol 2011 Fairclough Expphysiol.2010.053025

    31/31

    49 51 5248

    AO

    Exon 51 skipping

    49 5248

    Shorter functional mRNA

    TGA

    49 51 5248

    Exon 50 deletion in DMD mRNA with frameshift+PTC

    4948

    TGA

    49 50 51 5248

    Wild type dystrophin gene

    49 50 51 5248

    Full length functional mRNA

    TGA

    49 51 5248

    Exon 51 nonsense mutation in DMD

    49 5048

    mRNA with PTC

    50

    49 51 5248

    Read through of Exon 51

    TGA

    50

    Compoundsuppressing nonsense

    codons

    TGA

    49 50 51 5248

    Full length functional mRNA

    Figure 2

    51 52

    TGA

    TGA

    51 52

    A

    B

    C


Recommended