Invited review: Stem cells and muscle diseases: advances in cell therapy strategies

Neuropathology and Applied Neurobiology (2015), 41, 270–287 doi: 10.1111/nan.12198 Invited review: Stem cells and muscle diseases: advances in cell ...
Author: Chester Kelley
5 downloads 0 Views 299KB Size
Neuropathology and Applied Neurobiology (2015), 41, 270–287

doi: 10.1111/nan.12198

Invited review: Stem cells and muscle diseases: advances in cell therapy strategies E. Negroni, T. Gidaro, A. Bigot, G. S. Butler-Browne, V. Mouly and C. Trollet Institut de Myologie, CNRS FRE3617, UPMC Univ Paris 06, UM76, INSERM U974, Sorbonne Universités, 47 bd de l’Hôpital, Paris 75013, France

E. Negroni, T. Gidaro, A. Bigot, G. S. Butler-Browne, V. Mouly and C. Trollet (2015) Neuropathology and Applied Neurobiology 41, 270–287 Stem cells and muscle diseases: advances in cell therapy strategies Despite considerable progress to increase our understanding of muscle genetics, pathophysiology, molecular and cellular partners involved in muscular dystrophies and muscle ageing, there is still a crucial need for effective treatments to counteract muscle degeneration and muscle wasting in such conditions. This review focuses on cellbased therapy for muscle diseases. We give an overview of the different parameters that have to be taken into account in such a therapeutic strategy, including the influence of muscle ageing, cell proliferation and migra-

tion capacities, as well as the translation of preclinical results in rodent into human clinical approaches. We describe recent advances in different types of human myogenic stem cells, with a particular emphasis on myoblasts but also on other candidate cells described so far [CD133+ cells, aldehyde dehydrogenase-positive cells (ALDH+), muscle-derived stem cells (MuStem), embryonic stem cells (ES) and induced pluripotent stem cells (iPS)]. Finally, we provide an update of ongoing clinical trials using cell therapy strategies.

Keywords: cell therapy, muscle ageing, muscular dystrophy, myoblast, satellite cells, transplantation

Muscle diseases and therapies

Skeletal muscle Skeletal muscles constitute 40–50% of the body mass in adult human and encompass a broad range of different muscles. Skeletal muscle is a highly specialized tissue required for many physiological processes including locomotion, maintenance of posture, as well as metabolic activity. Skeletal muscle is able to tolerate chronic mechanical and physiological stresses throughout life to maintain proper contractile function. Skeletal muscle is composed of contractile units called muscle fibres bundled together and attached to the skeleton by tendons. Muscle Correspondence: Capucine Trollet, Myology Research Center, UMRS 974 UPMC – INSERM – FRE 3617 CNRS – AIM, 47, bld de l’hôpital – G.H. Pitié-Salpétrière – Bâtiment Babinski, 75651 Paris cedex 13, France. Tel: +33 01 4216 5715; Fax: +33 01 4216 5700; E-mail: [email protected]

270

fibres are post-mitotic multinucleated cells, surrounded by a specialized plasma membrane – the sarcolemma – and by a layer of extracellular matrix known as the basement membrane, which is composed of both an internal basal lamina and an external reticular lamina. The basal lamina associates closely with the sarcolemma providing a protective niche in which muscle precursors – called satellite cells (SC) [1] – reside. Muscle SC have a remarkable capacity to self renew and are also responsible for skeletal muscle growth and repair. Repair is crucial in conditions of muscle wasting including muscular dystrophies and muscle ageing. SC represent the adult stem cell of skeletal muscle [2]. Normally quiescent, in response to muscle damage, SC are able to activate, proliferate and fuse to repair the gap created in a myofibre by segmental necrosis or form new multinucleated myofibres. Identified in early studies on the basis of their position, beneath the basal lamina, by electron microscopy, SC are today identified by numerous © 2014 British Neuropathological Society

Advances in cell therapy strategies

271

Table 1. Muscle satellite cell markers Satellite cell state

Transcription factors Pax7 Myf5 MyoD Cell membrane proteins M-cadherin Caveolin 1 Syndecan 3 and 4 c-met CD34 CD56 CXCR4 a7integrin/B1integrin Cytoskeleton proteins Desmin Nestin

Detected in

Quiescent

Activation

Proliferation

Mouse

Human

+ − −

+ + +

+ + +

✓ ✓ ✓

✓ ? ✓

+ + + + + + + +

+ + + + + + ? +

+ + + + − + ? ?

✓ ✓ ✓ ✓ ✓ NA ✓ ✓

✓ ? ? ? ? ✓ ? ?

− +

+ +

+ ?

✓ ✓

✓ ?

NA, non applicable.

protein markers [3], allowing their identification in different cell state (quiescence, activation and proliferation) in mice and in humans (Table 1).

Satellite cells and muscle ageing Ageing results from the lifelong accumulation of subtle damages, caused by an exposure to biological and biochemical stresses. One of the detrimental hallmarks of muscle ageing is the decreased regenerative capacity after trauma or surgery, thus leading to longer immobilization, loss of muscle mass, increased sarcopenia and frailty. Age-related muscle decline can be exacerbated in patients with late onset degenerative disorders [4]. It is therefore essential to have a better understanding of the mechanisms of SC ageing to adapt therapeutic strategies, especially for late onset muscle diseases. The regenerative capacity of skeletal muscle is dependent on SC number and function, that is the ability of SC to activate, proliferate and differentiate. In mouse the decline in muscle regenerative capacity has been partly attributed to extrinsic environmental influences and diverse intrinsic potential of the SC themselves [5]. When old murine SC are exposed to a young environment or to growth factors, their capacity to proliferate and differentiate is partly restored [6–9], suggesting an influence of the muscle environment on the myogenic potency of old SC. In favour of a role for the environment, a recent © 2014 British Neuropathological Society

study underlines the function of oxytocin, a circulating hormone, which declines with age. A systemic administration of oxytocin improves muscle regeneration by enhancing activation – proliferation of aged SC [10]. One of the factors suggested to play a role in the decrease of regenerative capacity with age is the decrease in the number of resident stem cells. In humans, the number of SC decreases with age, from 4% to 5% in young adult subjects (∼20–30 years old) to 1% or less in old subjects (∼60–80 years old) [11,12]. SC depletion could be explained by a decrease in the ability of SC to return to quiescence to restore the pool of stem cells during the regeneration process or by an inability of SC to retain a quiescent state under homeostatic conditions. For example, an increase of fibroblast growth factor 2 (FGF2) from aged muscle fibres under homeostatic conditions leads to the loss of quiescence and depletion of SC [13], underlining the important role of the microenvironment in the regulation of SC function and maintenance of the stem cell pool. In a recent study, SousaVictor et al. demonstrated that the regenerative capacity of SC in geriatric mice cannot be restored after transplantation in young mice, as in geriatric mice, SC loose reversible quiescence and switch into a presenescence state. After injury, geriatric SC are therefore not able to participate to muscle regeneration and enter into a senescence state caused by derepression of the cell cycle inhibitor p16 [14]. A high level of p16 and senescent NAN 2015; 41: 270–287

272

E. Negroni et al.

markers associated with the elevated activity of both the p38α and p38β pathways were also described in aged mouse SC [15]. In addition, comparison between quiescent SC from young and old mice show that epigenetic changes accumulate with ageing and are linked to functional defects in aged SC [16]. All these results indicate that during homeostasis in the mouse, both the local environment as well as the micro-environment – that is niche of SC – play essential roles in SC regulation. This suggests that during murine regeneration the intrinsic defect present in the aged SC cannot be entirely corrected by a permissive environment. Several studies are working on improving the regeneration capacity of aged SC in mouse models, as an example, a rejuvenation of aged mouse SC was made by acting on p38α and p38β pathways in association with culture of SC on soft hydrogel substrates to develop autologous muscle cell transplantation [15]. These are important considerations that have to be kept in mind for the development of efficient cell therapy procedures. However, the situation differs in humans where very little differences were observed in proliferative capacity and in initial telomere length in SC isolated from adults with various ages, including elderly subjects [17]. This suggests that cell therapy can be a therapeutic option even in elderly subjects, as long as the SC have not exhausted their proliferative potential due to either a dystrophic process or extensive expansion in vitro and are still able to participate in the host’s regeneration, taking into account the difference in scale from mouse to human [18]. Although more work on human SC ageing needs to be developed in the future, one can already consider that whenever possible, e.g. in muscular dystrophies, the earlier the therapy starts, the highest chance of success it will have.

Muscular dystrophies Muscle tissue has more than 80 associated monogenic pathologies. Among them, muscular dystrophies (MDs) are a heterogeneous group of muscle diseases clinically characterized by progressive muscle wasting and weakness, with a wide clinical presentation and severity [19]. MDs have been classified according to age of onset (perinatal or adults), mode of inheritance (X-linked, autosomal recessive or dominant) and the specific muscle groups initially affected (limb-girdle, proximal, distal and facial muscles). Over the past years, more than 30 genetically distinct types of MDs have been identified [20,21] (see © 2014 British Neuropathological Society

http://www.musclegenetable.fr/ for an updated table of genetic neuromuscular disorders). Mutated genes involved in the development of MD encode a broad range of proteins located in the extracellular matrix (collagen and laminin), at the plasma membrane (dystrophin, dystroglycan, sarcoglycan, integrins, dysferlin), in the cytosol [fukutin related protein (FKRP), calpain 3, myotonic dystrophy type 1 (DM1) protein kinase (DMPK) and desmin], at the sarcomere (titin) and in the nucleus [laminAC, poly(A)-binding protein nuclear 1 (PABPN1) and emerin] of striated muscle cells. Interestingly, these proteins are not always muscle specific and are often ubiquitous: for example, the nuclear membrane protein emerin involved in Emery Dreifuss muscular dystrophy, the ubiquitous PABPN1 protein in oculopharyngeal muscular dystrophy (OPMD) or the dysferlin in limb-girdle muscular dystrophy. For many of these diseases, the pathophysiological mechanisms leading from the genetic mutation to the development of the dystrophic features are still unknown. In this section, we summarize the main muscular dystrophies in children and adults: a classification based on the pattern of muscle weakness and a multiorgan implication is proposed (Table 2). Duchenne muscular dystrophy (DMD) is the most common form of muscular dystrophy. DMD is an X-linked genetic disease affecting 1 in 3500 male births. DMD is caused by mutations in the dystrophin gene. Dystrophin is responsible for the maintenance of muscle fibre integrity, mediation of cytoplasmic signalling cascades and muscle functions [22–24]. Ambulation is usually lost between 10 and 12 years of age, and pulmonary insufficiency associated with cardiac involvement is the most frequent cause of death. In Becker muscular dystrophy, the allelic form of DMD, the distribution of muscle wasting and weakness is somehow similar to that in DMD, but the age of onset is around 8 years, and the course of the disease is more benign. Emery–Dreifuss muscular dystrophy (EDMD) is characterized by a triad of manifestations: i) early contractures of the Achilles tendons, elbows and posterior cervical muscles; ii) slowly progressive muscle wasting and weakness with a humeroperoneal distribution; iii) cardiomyopathy that usually presents as cardiac conduction defects, and starts by 30 years of age [25]. The X-linked form of EDMD is caused by mutations of the STA gene at Xq28, which encodes the nuclear membrane protein emerin [26]. The autosomal dominant type of this disorder is NAN 2015; 41: 270–287

Advances in cell therapy strategies

273

Table 2. Classification of muscular dystrophies

MD

Mode of inheritance

Age of onset

CPK

Childhood Adult

Very elevated Elevated

LGMD

X-linked X-linked or AD/AR AD/AR

Childhood Adult

Elevated

FSHD

AD

Childhood Adult

DM1

AD

OPMD

AD

Congenital Childhood Adult Late onset Adult

Normal or slightly elevated Normal or slightly elevated

DMD EDMD

Normal or slightly elevated

Weakness skeletal muscular distribution

Cardiac muscle involvement

Respiratory muscle involvement

Other systemic involvement

Lower limb-girdle Humero peronea distribution Upper and lower limb-girdle Facial and upper and lower limb-girdle

Yes Yes

Yes Possible

Brain* No

Possible

Possible

No

No

No

Retinal disease Hear

Distal or limb-girdle

Yes

Yes

Endocrine Brain Cataracts

Eyelid/pharyngeal

No

No

No

*Depending on exon type mutation. AD, autosomal dominant; AR, autosomal recessive; CPK, creatine phosphokinase.

clinically very similar but is caused by mutations of the LMNA gene at 1q21. This gene encodes lamins A and C, which make up part of the nuclear lamina [27]. Limb-girdle muscular dystrophy (LGMD) is typically an inherited disorder, with a dominant or recessive, genetic defect. LGMDs are mainly characterized by weakness of the proximal limb-girdle muscles, associated to myoglobinuria, pain, myotonia, cardiomyopathy, elevated serum creatine kinase (CK) and rippling muscles. To date, at least 20 genetically different types of LGMD have been identified, which show a great clinical and genetic heterogeneity [28]. The most common LGMD forms result from sarcolemmal adhesion complex defects, as sarcoglycans in LGMD type2 C-F, but also other sarcolemmal molecules, such as caveolin-3 in LGMD type1C and dysferlin in LGMD type 2B, or nuclear membrane, as lamin A/C in LGMD type 1B. Facioscapulohumeral muscular dystrophy (FSHD) is transmitted by an autosomal dominant inheritance associated with a contraction of the tandem 3.3-kb D4Z4 repeat unit at chromosome 4q35 [29]. Each D4Z4 repeat contains a copy of the DUX4 gene that potentially results in a toxic gain of function when expressed and appears to contribute to the pathogenesis of FSHD [30]. In FSHD, facial and shoulder girdle are the muscle groups that are mainly affected. Later, foot extensors and pelvic-girdle muscles become involved. © 2014 British Neuropathological Society

DM1, also known as Steinert’s disease, is a progressive muscle degeneration disease leading to disabling weakness and wasting with myotonia, in combination with multisystemic involvement (cataracts, cognitive impairments, cardiac and pulmonary dysfunctions). DM1 is the most common form of adult-onset neuromuscular disorder with a prevalence ranging from 2 to 14 per 100,000population worldwide. DM1 is caused by a (CTG)n microsatellite repeat expansion in the untranslated 3′ region of the DMPK gene on chromosome 19q13.3 [31]. This unstable expansion segregates as an autosomal dominant trait with incomplete penetrance and greatly variable expressivity. Four clinical forms of DM1 are recognized according to (CTG)n repeat number inversely proportional to the age of onset: congenital DM1 (>1000 CTG), childhood DM1 (21 8–17 26 8–12 18–75 18–65 6–14 None none None

− − −

2 ml divided into 26 small depots

Injection 0.5 ml each with prior electric stimulation Single injection, 1–2 ml

12 injections into an area of 10 cm2 with prior scarification

Grid of 55–60 points 1 cm apart, 0.1 ml myoblast mixture Zone of injection of 1 cm3, 25 parallel injections 5 mm grid device, 100–200 injections, 2–21 cm2 surface, in three sessions, with 2 days intervals Three parallel injections at 1 mm interdistance

5.5 ml with 55 sites around 5 tracks

80–100 sites in a grid (4 × 6 cm) spaced 5 mm apart, 2.5 cm depth 100 μl per injection, 55 sites at 5 mm intervals, 2.5 cm depth 2 or 3 injection site, 30 million cells/ml

Urethral sphincter

urethral sphincter

External anal sphincter

TA

Pharyngeal

Abductor digiti minimi

Gastrocnemius, biceps brachii, thenar eminence

12 months follow-up, safe, well-tolerated, quality of life improved 1,2,3,6and 12 months, quality of life improved in 50% of patients; 10 of 12 patients improved Follow-up for 6 months, 23.7% cured and 52.6% improvement at 6 months

Gastrocnemius was Dys+ at 18 months (PCR and IHC), very slightly the biceps brachii (PCR at 14 months). Thenar eminence was not analysed. Safety test: neither local nor systemic adverse effects were reported. Increase vascularization into 4 out of 5 treated muscles. Safe procedure, good tolerability, cell dose effect

8/9 patients Dys+ (IHC)

TA

TA

No Dys+ at 1 year follow-up but 3/8 patients showed improved strength Improved strengths (12–31%) in wrist extension at 6 months; very slight increase in Dys expression 3/4 patients Dys+ (IHC and WB) No Dys+ 1/12 patients Dys+ (IHC) 3/10 patients Dys+ (PCR) at 1 month; 1/6 at 6 months 0/3 Dys+ (IHC) at 6 months

Biceps brachii

[131]

[129]

− [130]



[128]

[127]

[67]

[126]

[125]

[121] [122] [123] [124]

[120]

[119]

[118]

[117]

Ref.

Myoblast Myoblast Myoblast Myoblast Myoblast Myoblast Myoblast Myoblast Myoblast Myoblast Myoblast CD133+ Myoblast Myoblast HLA-identical allogenic mesoangiblasts Myoblast Myoblast Myoblast

Cell type

7/9 patients (TA) Dys+ (IHC/WB) + 4/9 showed improved strength at 4 months 3/8 patients Dys+ (PCR) at 1 month

A A A

H H H H* H H H H H H H A A A H

Autologous/ heterologous

Extensor carpi radialis, biceps brachii Biceps brachii, TA TA Biceps brachii TA

TA, biceps brachii, and/or extensor carpi radialis longus TA

Outcomes

None None Cyclophosphamide for 6–12 months None None Cyclosporine (5 days before and 6 months after) Cyclosporine Cyclosporine for 7 days Cyclosporine 2 months before and 1 year after Tacrolimus (7 days before and maintained throughout the study) Tacrolimus (7 days before and maintained throughout the study) None None None

− − + − − + 6 patients + 6 patients + + + + − − − +

Recipient muscle

Immunosuppression regime

Immunosuppression

Mode of injection

Patients’ age

*Donor was the monozygotic twin of symptomatic carrier. A, autologous; BMD, Becker muscular dystrophy; DMD, Duchenne muscular dystrophy; FSHD, Facioscapulohumeral dystrophy; H, heterologous; IHC, immunohistochemistry; im, intramuscular; ia, intraarterial; WB, Western blot; EBD, extensor digitorum brevis; OPMD, oculopharyngeal muscular dystrophy; SCM, sterno-cleido-mastoidien; TA, tibialis anterior.

Biceps brachii

Deltoid

1993

0.5–1g

Anal incontinence Stress urinary incontinence Stress urinary incontinence

1993



Biceps

1992

Donor muscle

Year

1992

Muscle biopsy weight

Frudinger Sebe Blaganje

2009 2011 2013

DMD DMD DMD DMD DMD DMD DMD DMD BMD DMD DMD DMD OPMD FSHD DMD

Huard Gussoni Karpati Tremblay Tremblay Morandi Mendell Miller Neumeyer Skuk Skuk Torrente Perie

1992 1992 1993 1993 1993 1995 1995 1997 1998 2004–2006 2007 2007 2013 Ongoing Ongoing

Disease

First author

Year

Table 3. Muscle cell therapy clinical studies (cardiac muscle target is not included)

Advances in cell therapy strategies 281

NAN 2015; 41: 270–287

282

E. Negroni et al.

bottlenecks have been rapidly identified in preclinical and clinical studies. The lessons we learned from over 25 years of research in this field tell us that each pathological situation has to be considered individually and that therapeutic strategies will have to be adapted to each unique situation. For example, the lack of clinical benefit of myoblast implantation in DMD patients should be compared with the clinical improvement observed in OPMD patients after implantation of the same cell type. If limited targets – such as those in OPMD – can already be treated by cell therapy, extended treatment – such as those required for other muscle dystrophies including DMD – will require further research efforts. Over the past years, many different cell types with a myogenic potential were described. A particular effort will have to be made on the research conducted to find cell types that can be systemically delivered. In addition, we should synergize the different therapeutic strategies developed in the past to make profit of all the progresses made in gene, cell or pharmacotherapy. As an example, autologous cell transplantation may require gene correction of the cells before implantation. Pharmacological treatments of the recipient muscle, to improve muscle function, may also increase the overall efficiency of gene or cell therapy strategies. A combination of all these tools will be instrumental in fighting these devastating diseases.

Acknowledgements The authors acknowledge Jean-Thomas Vilquin for fruitful discussions. This study was supported by grants from AFM (Association Française contre les Myopathies, OPMD Network Research Program 15123), MYOAGE (contract HEALTH-F2-2009-223576) from the 7th FP, the ANR Genopath-INAFIB, Fondation de l’avenir (project ET1622), INSERM, CNRS and Université Pierre et Marie Curie.

Authors contribution EN, TG, AB, VM, GBB and CT contributed equally to the writing of the manuscript and generation of the tables.

Conflicts of interest None.

References 1 Mauro A. Satellite cell of skeletal muscle fibers. J Biophys Biochem Cytol 1961; 9: 493–5 © 2014 British Neuropathological Society

2 Morgan JE, Partridge TA. Muscle satellite cells. Int J Biochem Cell Biol 2003; 35: 1151–6 3 Boldrin L, Muntoni F, Morgan JE. Are human and mouse satellite cells really the same? J Histochem Cytochem 2010; 58: 941–55 4 Kirkwood TB, Austad SN. Why do we age? Nature 2000; 408: 233–8 5 Collins CA, Olsen I, Zammit PS, Heslop L, Petrie A, Partridge TA, Morgan JE. Stem cell function, selfrenewal, and behavioral heterogeneity of cells from the adult muscle satellite cell niche. Cell 2005; 122: 289– 301 6 Brack AS, Conboy MJ, Roy S, Lee M, Kuo CJ, Keller C, Rando TA. Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science 2007; 317: 807–10 7 Carlson BM, Faulkner JA. Muscle transplantation between young and old rats: age of host determines recovery. Am J Physiol 1989; 256: C1262–6 8 Collins CA, Zammit PS, Ruiz AP, Morgan JE, Partridge TA. A population of myogenic stem cells that survives skeletal muscle aging. Stem Cells 2007; 25: 885–94 9 Conboy IM, Rando TA. Heterochronic parabiosis for the study of the effects of aging on stem cells and their niches. Cell Cycle 2013; 11: 2260–7 10 Elabd C, Cousin W, Upadhyayula P, Chen RY, Chooljian MS, Li J, Kung S, Jiang KP, Conboy IM. Oxytocin is an age-specific circulating hormone that is necessary for muscle maintenance and regeneration. Nat Commun 2014; 5: 4082–93 11 Renault V, Thornell LE, Eriksson PO, Butler-Browne G, Mouly V. Regenerative potential of human skeletal muscle during aging. Aging Cell 2002; 1: 132–9 12 Kadi F, Charifi N, Denis C, Lexell J. Satellite cells and myonuclei in young and elderly women and men. Muscle Nerve 2004; 29: 120–7 13 Chakkalakal JV, Jones KM, Basson MA, Brack AS. The aged niche disrupts muscle stem cell quiescence. Nature 2012; 490: 355–60 14 Sousa-Victor P, Gutarra S, Garcia-Prat L, Rodriguez-Ubreva J, Ortet L, Ruiz-Bonilla V, Jardi M, Ballestar E, Gonzalez S, Serrano AL, Perdiguero E, Munoz-Canoves P. Geriatric muscle stem cells switch reversible quiescence into senescence. Nature 2014; 506: 316–21 15 Cosgrove BD, Gilbert PM, Porpiglia E, Mourkioti F, Lee SP, Corbel SY, Llewellyn ME, Delp SL, Blau HM. Rejuvenation of the muscle stem cell population restores strength to injured aged muscles. Nat Med 2014; 20: 255–64 16 Liu L, Cheung TH, Charville GW, Hurgo BM, Leavitt T, Shih J, Brunet A, Rando TA. Chromatin modifications as determinants of muscle stem cell quiescence and chronological aging. Cell Rep 2014; 4: 189–204 17 Decary S, Mouly V, Hamida CB, Sautet A, Barbet JP, Butler-Browne GS. Replicative potential and telomere NAN 2015; 41: 270–287

Advances in cell therapy strategies

18

19 20

21

22

23

24

25 26

27

28 29

30

31

length in human skeletal muscle: implications for satellite cell-mediated gene therapy. Hum Gene Ther 1997; 8: 1429–38 Partridge TA. The mdx mouse model as a surrogate for Duchenne muscular dystrophy. FEBS J 2013; 280: 4177–86 Emery AE. The muscular dystrophies. Lancet 2002; 359: 687–95 Bushby K, Lochmuller H, Lynn S, Straub V. Interventions for muscular dystrophy: molecular medicines entering the clinic. Lancet 2009; 374: 1849–56 Kaplan JC. Gene table of monogenic neuromuscular disorders (nuclear genome only) Vol 19. No 1 January 2009. Neuromuscul Disord 2009; 19: 77–98 Kumar A, Khandelwal N, Malya R, Reid MB, Boriek AM. Loss of dystrophin causes aberrant mechanotransduction in skeletal muscle fibers. FASEB J 2004; 18: 102–13 Seno MM, Trollet C, Athanasopoulos T, Graham IR, Hu P, Dickson G. Transcriptomic analysis of dystrophin RNAi knockdown reveals a central role for dystrophin in muscle differentiation and contractile apparatus organization. BMC Genomics 2010; 11: 345 Sweeney HL, Barton ER. The dystrophin-associated glycoprotein complex: what parts can you do without? Proc Natl Acad Sci U S A 2000; 97: 13464–6 Emery AE. Emery-Dreifuss syndrome. J Med Genet 1989; 26: 637–41 Bione S, Maestrini E, Rivella S, Mancini M, Regis S, Romeo G, Toniolo D. Identification of a novel X-linked gene responsible for Emery-Dreifuss muscular dystrophy. Nat Genet 1994; 8: 323–7 Bonne G, Di Barletta MR, Varnous S, Becane HM, Hammouda EH, Merlini L, Muntoni F, Greenberg CR, Gary F, Urtizberea JA, Duboc D, Fardeau M, Toniolo D, Schwartz K. Mutations in the gene encoding lamin A/C cause autosomal dominant Emery-Dreifuss muscular dystrophy. Nat Genet 1999; 21: 285– 8 Bushby K. The limb-girdle muscular dystrophies. Eur J Paediatr Neurol 2001; 5: 213–14 Wijmenga C, Frants RR, Brouwer OF, Moerer P, Weber JL, Padberg GW. Location of facioscapulohumeral muscular dystrophy gene on chromosome 4. Lancet 1990; 336: 651–3 Lemmers RJ, van der Vliet PJ, Klooster R, Sacconi S, Camano P, Dauwerse JG, Snider L, Straasheijm KR, van Ommen GJ, Padberg GW, Miller DG, Tapscott SJ, Tawil R, Frants RR, van der Maarel SM. A unifying genetic model for facioscapulohumeral muscular dystrophy. Science 2010; 329: 1650–3 Mahadevan MS, Yadava RS, Yu Q, Balijepalli S, Frenzel-McCardell CD, Bourne TD, Phillips LH. Reversible model of RNA toxicity and cardiac conduction defects in myotonic dystrophy. Nat Genet 2006; 38: 1066–70

© 2014 British Neuropathological Society

283

32 Harley HG, Rundle SA, MacMillan JC, Myring J, Brook JD, Crow S, Reardon W, Fenton I, Shaw DJ, Harper PS. Size of the unstable CTG repeat sequence in relation to phenotype and parental transmission in myotonic dystrophy. Am J Hum Genet 1993; 52: 1164–74 33 Brais B, Bouchard JP, Xie YG, Rochefort DL, Chretien N, Tome FM, Lafreniere RG, Rommens JM, Uyama E, Nohira O, Blumen S, Korczyn AD, Heutink P, Mathieu J, Duranceau A, Codere F, Fardeau M, Rouleau GA. Short GCG expansions in the PABP2 gene cause oculopharyngeal muscular dystrophy. Nat Genet 1998; 18: 164– 7 34 Banerjee A, Apponi LH, Pavlath GK, Corbett AH. PABPN1: molecular function and muscle disease. FEBS J 2013; 280: 4230–50 35 Gidaro T, Negroni E, Perie S, Mirabella M, Laine J, Lacau St Guily J, Butler-Browne G, Mouly V, Trollet C. Atrophy, fibrosis, and increased PAX7-positive cells in pharyngeal muscles of oculopharyngeal muscular dystrophy patients. J Neuropathol Exp Neurol 2013; 72: 234–43 36 Khurana TS, Davies KE. Pharmacological strategies for muscular dystrophy. Nat Rev Drug Discov 2003; 2: 379–90 37 Mozzetta C, Minetti G, Puri PL. Regenerative pharmacology in the treatment of genetic diseases: the paradigm of muscular dystrophy. Int J Biochem Cell Biol 2009; 41: 701–10 38 Koo T, Wood MJ. Clinical trials using antisense oligonucleotides in Duchenne muscular dystrophy. Hum Gene Ther 2013; 24: 479–88 39 Trollet C, Athanasopoulos T, Popplewell L, Malerba A, Dickson G. Gene therapy for muscular dystrophy: current progress and future prospects. Expert Opin Biol Ther 2009; 9: 849–66 40 Nayak S, Herzog RW. Progress and prospects: immune responses to viral vectors. Gene Ther 2010; 17: 295– 304 41 Odom GL, Gregorevic P, Chamberlain JS. Viral-mediated gene therapy for the muscular dystrophies: successes, limitations and recent advances. Biochim Biophys Acta 2007; 1772: 243–62 42 Tedesco FS, Dellavalle A, Diaz-Manera J, Messina G, Cossu G. Repairing skeletal muscle: regenerative potential of skeletal muscle stem cells. J Clin Invest 2010; 120: 11–19 43 Negroni E, Vallese D, Vilquin JT, Butler-Browne G, Mouly V, Trollet C. Current advances in cell therapy strategies for muscular dystrophies. Expert Opin Biol Ther 2011; 11: 157–76 44 Tedesco FS, Cossu G. Stem cell therapies for muscle disorders. Curr Opin Neurol 2012; 25: 597–603 45 Zammit PS, Partridge TA, Yablonka-Reuveni Z. The skeletal muscle satellite cell: the stem cell that came in from the cold. J Histochem Cytochem 2006; 54: 1177–91 NAN 2015; 41: 270–287

284

E. Negroni et al.

46 Partridge TA, Morgan JE, Coulton GR, Hoffman EP, Kunkel LM. Conversion of mdx myofibres from dystrophin-negative to -positive by injection of normal myoblasts. Nature 1989; 337: 176–9 47 Kinoshita I, Vilquin JT, Guerette B, Asselin I, Roy R, Tremblay JP. Very efficient myoblast allotransplantation in mice under FK506 immunosuppression. Muscle Nerve 1994; 17: 1407–15 48 Kinoshita I, Huard J, Tremblay JP. Utilization of myoblasts from transgenic mice to evaluate the efficacy of myoblast transplantation. Muscle Nerve 1994; 17: 975–80 49 Huard J, Verreault S, Roy R, Tremblay M, Tremblay JP. High efficiency of muscle regeneration after human myoblast clone transplantation in SCID mice. J Clin Invest 1994; 93: 586–99 50 Huard J, Tremblay G, Verreault S, Labrecque C, Tremblay JP. Utilization of an antibody specific for human dystrophin to follow myoblast transplantation in nude mice. Cell Transplant 1993; 2: 113–18 51 Skuk D, Goulet M, Tremblay JP. Intramuscular transplantation of myogenic cells in primates: importance of needle size, cell number, and injection volume. Cell Transplant 2014; 23: 13–25 52 Beauchamp J, Morgan J, Pagel C, Partridge T. Dynamics of myoblast transplantation reveal a discrete minority of precursors with stem cell-like properties as the myogenic source. J Cell Biol 1999; 144: 1113–22 53 Skuk D, Caron N, Goulet M, Roy B, Espinosa F, Tremblay JP. Dynamics of the early immune cellular reactions after myogenic cell transplantation. Cell Transplant 2002; 11: 671–81 54 Skuk D, Caron NJ, Goulet M, Roy B, Tremblay JP. Resetting the problem of cell death following muscle-derived cell transplantation: detection, dynamics and mechanisms. J Neuropathol Exp Neurol 2003; 62: 951–67 55 Vilquin JT, Catelain C, Vauchez K. Cell therapy for muscular dystrophies: advances and challenges. Curr Opin Organ Transplant 2011; 16: 640–9 56 Vallese D, Negroni E, Duguez S, Ferry A, Trollet C, Aamiri A, Vosshenrich CA, Fuchtbauer EM, Di Santo JP, Vitiello L, Butler-Browne G, Mouly V. The Rag2(-)Il2rb()Dmd(-) mouse: a novel dystrophic and immunodeficient model to assess innovating therapeutic strategies for muscular dystrophies. Mol Ther 2013; 21: 1950–7 57 Cooper RN, Irintchev A, Di Santo JP, Zweyer M, Morgan JE, Partridge TA, Butler-Browne GS, Mouly V, Wernig A. A new immunodeficient mouse model for human myoblast transplantation. Hum Gene Ther 2001; 12: 823–31 58 Arpke RW, Darabi R, Mader TL, Zhang Y, Toyama A, Lonetree CL, Nash N, Lowe DA, Perlingeiro RC, Kyba M. A new immuno-, dystrophin-deficient model, the NSGmdx(4Cv) mouse, provides evidence for functional improvement following allogeneic satellite cell transplantation. Stem Cells 2013; 31: 1611–20 © 2014 British Neuropathological Society

59 Riederer I, Negroni E, Bencze M, Wolff A, Aamiri A, Di Santo JP, Silva-Barbosa SD, Butler-Browne G, Savino W, Mouly V. Slowing down differentiation of engrafted human myoblasts into immunodeficient mice correlates with increased proliferation and migration. Mol Ther 2012; 20: 146–54 60 Lipton BH, Schultz E. Developmental fate of skeletal muscle satellite cells. Science 1979; 205: 1292–4 61 Rando TA, Pavlath GK, Blau HM. The fate of myoblasts following transplantation into mature muscle. Exp Cell Res 1995; 220: 383–9 62 Quenneville SP, Chapdelaine P, Skuk D, Paradis M, Goulet M, Rousseau J, Xiao X, Garcia L, Tremblay JP. Autologous transplantation of muscle precursor cells modified with a lentivirus for muscular dystrophy: human cells and primate models. Mol Ther 2007; 15: 431–8 63 Skuk D, Goulet M, Tremblay JP. Transplanted myoblasts can migrate several millimeters to fuse with damaged myofibers in nonhuman primate skeletal muscle. J Neuropathol Exp Neurol 2011; 70: 770–8 64 Skuk D, Furling D, Bouchard JP, Goulet M, Roy B, Lacroix Y, Vilquin JT, Tremblay JP, Puymirat J. Transplantation of human myoblasts in SCID mice as a potential muscular model for myotonic dystrophy. J Neuropathol Exp Neurol 1999; 58: 921–31 65 Morgan JE, Pagel CN, Sherratt T, Partridge TA. Longterm persistence and migration of myogenic cells injected into pre-irradiated muscles of mdx mice. J Neurol Sci 1993; 115: 191–200 66 Morgan JE, Hoffman EP, Partridge TA. Normal myogenic cells from newborn mice restore normal histology to degenerating muscles of the mdx mouse. J Cell Biol 1990; 111: 2437–49 67 Skuk D, Goulet M, Roy B, Piette V, Cote CH, Chapdelaine P, Hogrel JY, Paradis M, Bouchard JP, Sylvain M, Lachance JG, Tremblay JP. First test of a ‘high-density injection’ protocol for myogenic cell transplantation throughout large volumes of muscles in a Duchenne muscular dystrophy patient: eighteen months followup. Neuromuscul Disord 2007; 17: 38–46 68 Skuk D, Goulet M, Roy B, Chapdelaine P, Bouchard JP, Roy R, Dugre FJ, Sylvain M, Lachance JG, Deschenes L, Senay H, Tremblay JP. Dystrophin expression in muscles of Duchenne muscular dystrophy patients after high-density injections of normal myogenic cells. J Neuropathol Exp Neurol 2006; 65: 371–86 69 Tanaka KK, Hall JK, Troy AA, Cornelison DD, Majka SM, Olwin BB. Syndecan-4-expressing muscle progenitor cells in the SP engraft as satellite cells during muscle regeneration. Cell Stem Cell 2009; 4: 217–25 70 Sacco A, Doyonnas R, Kraft P, Vitorovic S, Blau HM. Self-renewal and expansion of single transplanted muscle stem cells. Nature 2008; 456: 502–6 71 Montarras D, Morgan J, Collins C, Relaix F, Zaffran S, Cumano A, Partridge T, Buckingham M. Direct isolation NAN 2015; 41: 270–287

Advances in cell therapy strategies

72

73

74

75

76

77

78

79

80

81

82

of satellite cells for skeletal muscle regeneration. Science 2005; 309: 2064–7 Cerletti M, Shadrach JL, Jurga S, Sherwood R, Wagers AJ. Regulation and function of skeletal muscle stem cells. Cold Spring Harb Symp Quant Biol 2008; 73: 317–22 Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell 2006; 126: 677–89 Gilbert PM, Havenstrite KL, Magnusson KE, Sacco A, Leonardi NA, Kraft P, Nguyen NK, Thrun S, Lutolf MP, Blau HM. Substrate elasticity regulates skeletal muscle stem cell self-renewal in culture. Science 2010; 329: 1078–81 Yennek S, Burute M, Thery M, Tajbakhsh S. Cell adhesion geometry regulates non-random DNA segregation and asymmetric cell fates in mouse skeletal muscle stem cells. Cell Rep 2014; 7: 961–70 Bencze M, Negroni E, Vallese D, Yacoub-Youssef H, Chaouch S, Wolff A, Aamiri A, Di Santo JP, Chazaud B, Butler-Browne G, Savino W, Mouly V, Riederer I. Proinflammatory macrophages enhance the regenerative capacity of human myoblasts by modifying their kinetics of proliferation and differentiation. Mol Ther 2012; 20: 2168–79 Motohashi N, Uezumi A, Yada E, Fukada S, Fukushima K, Imaizumi K, Miyagoe-Suzuki Y, Takeda S. Muscle CD31(-) CD45(-) side population cells promote muscle regeneration by stimulating proliferation and migration of myoblasts. Am J Pathol 2008; 173: 781–91 Mozzetta C, Consalvi S, Saccone V, Tierney M, Diamantini A, Mitchell KJ, Marazzi G, Borsellino G, Battistini L, Sassoon D, Sacco A, Puri PL. Fibroadipogenic progenitors mediate the ability of HDAC inhibitors to promote regeneration in dystrophic muscles of young, but not old Mdx mice. EMBO Mol Med 2013; 5: 626–39 Joe AW, Yi L, Natarajan A, Le Grand F, So L, Wang J, Rudnicki MA, Rossi FM. Muscle injury activates resident fibro/adipogenic progenitors that facilitate myogenesis. Nat Cell Biol 2010; 12: 153–63 Zhang Y, King OD, Rahimov F, Jones TI, Ward CW, Kerr JP, Liu N, Emerson CP Jr, Kunkel LM, Partridge TA, Wagner KR. Human skeletal muscle xenograft as a new preclinical model for muscle disorders. Hum Mol Genet 2014; 23: 3180–8 Bentzinger CF, von Maltzahn J, Dumont NA, Stark DA, Wang YX, Nhan K, Frenette J, Cornelison DD, Rudnicki MA. Wnt7a stimulates myogenic stem cell motility and engraftment resulting in improved muscle strength. J Cell Biol 2014; 205: 97–111 Dellavalle A, Sampaolesi M, Tonlorenzi R, Tagliafico E, Sacchetti B, Perani L, Innocenzi A, Galvez BG, Messina G, Morosetti R, Li S, Belicchi M, Peretti G, Chamberlain JS, Wright WE, Torrente Y, Ferrari S, Bianco P, Cossu G. Pericytes of human skeletal muscle are myogenic

© 2014 British Neuropathological Society

83

84

85

86

87

88

89

90

91

92

285

precursors distinct from satellite cells. Nat Cell Biol 2007; 9: 255–67 Torrente Y, Belicchi M, Sampaolesi M, Pisati F, Meregalli M, D’Antona G, Tonlorenzi R, Porretti L, Gavina M, Mamchaoui K, Pellegrino MA, Furling D, Mouly V, Butler-Browne GS, Bottinelli R, Cossu G, Bresolin N. Human circulating AC133(+) stem cells restore dystrophin expression and ameliorate function in dystrophic skeletal muscle. J Clin Invest 2004; 114: 182–95 Shi M, Ishikawa M, Kamei N, Nakasa T, Adachi N, Deie M, Asahara T, Ochi M. Acceleration of skeletal muscle regeneration in a rat skeletal muscle injury model by local injection of human peripheral blood-derived CD133-positive cells. Stem Cells 2009; 27: 949–60 Negroni E, Riederer I, Chaouch S, Belicchi M, Razini P, Di Santo J, Torrente Y, Butler-Browne GS, Mouly V. In vivo myogenic potential of human CD133+ musclederived stem cells: a quantitative study. Mol Ther 2009; 17: 1771–8 Benchaouir R, Meregalli M, Farini A, D’Antona G, Belicchi M, Goyenvalle A, Battistelli M, Bresolin N, Bottinelli R, Garcia L, Torrente Y. Restoration of human dystrophin following transplantation of exon-skippingengineered DMD patient stem cells into dystrophic mice. Cell Stem Cell 2007; 1: 646–57 Vauchez K, Marolleau JP, Schmid M, Khattar P, Chapel A, Catelain C, Lecourt S, Larghero J, Fiszman M, Vilquin JT. Aldehyde dehydrogenase activity identifies a population of human skeletal muscle cells with high myogenic capacities. Mol Ther 2009; 17: 1948–58 Rouger K, Larcher T, Dubreil L, Deschamps JY, Le Guiner C, Jouvion G, Delorme B, Lieubeau B, Carlus M, Fornasari B, Theret M, Orlando P, Ledevin M, Zuber C, Leroux I, Deleau S, Guigand L, Testault I, Le Rumeur E, Fiszman M, Cherel Y. Systemic delivery of allogenic muscle stem cells induces long-term muscle repair and clinical efficacy in Duchenne muscular dystrophy dogs. Am J Pathol 2011; 179: 2501–18 De Angelis L, Berghella L, Coletta M, Lattanzi L, Zanchi M, Cusella-De Angelis MG, Ponzetto C, Cossu G. Skeletal myogenic progenitors originating from embryonic dorsal aorta coexpress endothelial and myogenic markers and contribute to postnatal muscle growth and regeneration. J Cell Biol 1999; 147: 869–78 Sampaolesi M, Torrente Y, Innocenzi A, Tonlorenzi R, D’Antona G, Pellegrino MA, Barresi R, Bresolin N, De Angelis MG, Campbell KP, Bottinelli R, Cossu G. Cell therapy of alpha-sarcoglycan null dystrophic mice through intra-arterial delivery of mesoangioblasts. Science 2003; 301: 487–92 Cossu G, Bianco P. Mesoangioblasts–vascular progenitors for extravascular mesodermal tissues. Curr Opin Genet Dev 2003; 13: 537–42 Sampaolesi M, Blot S, Antona D, Granger G, Tonlorenzi N, Innocenzi R, Mognol A, Thibaud P, Galvez JL, Barthelemy BG, Perani I, Mantero L, Guttinger S, NAN 2015; 41: 270–287

286

93

94

95

96

97

98

99

100

101 102

103

104

E. Negroni et al.

Pansarasa M, Rinaldi O, Cusella C, De Angelis MG, Torrente Y, Bordignon C, Bottinelli R, Cossu G. Mesoangioblast stem cells ameliorate muscle function in dystrophic dogs. Nature 2006; 444: 574–9 Noviello M, Tedesco FS, Bondanza A, Tonlorenzi R, Rosaria Carbone M, Gerli MF, Marktel S, Napolitano S, Cicalese MP, Ciceri F, Peretti G, Cossu G, Bonini C. Inflammation converts human mesoangioblasts into targets of alloreactive immune responses: implications for allogeneic cell therapy of DMD. Mol Ther 2014; 22: 1342–52 Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. Embryonic stem cell lines derived from human blastocysts. Science 1998; 282: 1145–7 Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007; 131: 861–72 Meissner A, Wernig M, Jaenisch R. Direct reprogramming of genetically unmodified fibroblasts into pluripotent stem cells. Nat Biotechnol 2007; 25: 1177–81 Bodnar MS, Meneses JJ, Rodriguez RT, Firpo MT. Propagation and maintenance of undifferentiated human embryonic stem cells. Stem Cells Dev 2004; 13: 243–53 Barberi T, Bradbury M, Dincer Z, Panagiotakos G, Socci ND, Studer L. Derivation of engraftable skeletal myoblasts from human embryonic stem cells. Nat Med 2007; 13: 642–8 Darabi R, Gehlbach K, Bachoo RM, Kamath S, Osawa M, Kamm KE, Kyba M, Perlingeiro RC. Functional skeletal muscle regeneration from differentiating embryonic stem cells. Nat Med 2008; 14: 134–43 Darabi R, Arpke RW, Irion S, Dimos JT, Grskovic M, Kyba M, Perlingeiro RC. Human ES- and iPS-derived myogenic progenitors restore dystrophin and improve contractility upon transplantation in dystrophic mice. Cell Stem Cell 2012; 10: 610–19 Chidgey AP, Boyd RL. Immune privilege for stem cells: not as simple as it looked. Cell Stem Cell 2008; 3: 357–8 Swijnenburg RJ, Tanaka M, Vogel H, Baker J, Kofidis T, Gunawan F, Lebl DR, Caffarelli AD, de Bruin JL, Fedoseyeva EV, Robbins RC. Embryonic stem cell immunogenicity increases upon differentiation after transplantation into ischemic myocardium. Circulation 2005; 112: I166–72 Yamanaka S, Li J, Kania G, Elliott S, Wersto RP, Van Eyk J, Wobus AM, Boheler KR. Pluripotency of embryonic stem cells. Cell Tissue Res 2008; 331: 5–22 Ichida JK, Blanchard J, Lam K, Son EY, Chung JE, Egli D, Loh KM, Carter AC, Di Giorgio FP, Koszka K, Huangfu D, Akutsu H, Liu DR, Rubin LL, Eggan K. A small-molecule inhibitor of tgf-Beta signaling replaces sox2 in reprogramming by inducing nanog. Cell Stem Cell 2009; 5: 491–503

© 2014 British Neuropathological Society

105 Kim D, Kim CH, Moon JI, Chung YG, Chang MY, Han BS, Ko S, Yang E, Cha KY, Lanza R, Kim KS. Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins. Cell Stem Cell 2009; 4: 472–6 106 Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S. Generation of mouse induced pluripotent stem cells without viral vectors. Science 2008; 322: 949–53 107 Judson RL, Babiarz JE, Venere M, Blelloch R. Embryonic stem cell-specific microRNAs promote induced pluripotency. Nat Biotechnol 2009; 27: 459–61 108 Park IH, Arora N, Huo H, Maherali N, Ahfeldt T, Shimamura A, Lensch MW, Cowan C, Hochedlinger K, Daley GQ. Disease-specific induced pluripotent stem cells. Cell 2008; 134: 877–86 109 Yamanaka S. Patient-specific pluripotent stem cells become even more accessible. Cell Stem Cell 2010; 7: 1–2 110 Mizuno Y, Chang H, Umeda K, Niwa A, Iwasa T, Awaya T, Fukada S, Yamamoto H, Yamanaka S, Nakahata T, Heike T. Generation of skeletal muscle stem/progenitor cells from murine induced pluripotent stem cells. FASEB J 2010; 24: 2245–53 111 Darabi R, Baik J, Clee M, Kyba M, Tupler R, Perlingeiro RC. Engraftment of embryonic stem cell-derived myogenic progenitors in a dominant model of muscular dystrophy. Exp Neurol 2009; 220: 212–16 112 Tedesco FS, Gerli MF, Perani L, Benedetti S, Ungaro F, Cassano M, Antonini S, Tagliafico E, Artusi V, Longa E, Tonlorenzi R, Ragazzi M, Calderazzi G, Hoshiya H, Cappellari O, Mora M, Schoser B, Schneiderat P, Oshimura M, Bottinelli R, Sampaolesi M, Torrente Y, Broccoli V, Cossu G. Transplantation of genetically corrected human iPSC-derived progenitors in mice with limb-girdle muscular dystrophy. Sci Transl Med 2012; 4: 140ra89 113 Watt DJ, Morgan JE, Partridge TA. Use of mononuclear precursor cells to insert allogeneic genes into growing mouse muscles. Muscle Nerve 1984; 7: 741–50 114 Watt DJ, Lambert K, Morgan JE, Partridge TA, Sloper JC. Incorporation of donor muscle precursor cells into an area of muscle regeneration in the host mouse. J Neurol Sci 1982; 57: 319–31 115 Morgan JE, Watt DJ, Sloper JC, Partridge TA. Partial correction of an inherited biochemical defect of skeletal muscle by grafts of normal muscle precursor cells. J Neurol Sci 1988; 86: 137–47 116 Partridge TA. Invited review: myoblast transfer: a possible therapy for inherited myopathies? Muscle Nerve 1991; 14: 197–212 117 Huard J, Roy R, Bouchard JP, Malouin F, Richards CL, Tremblay JP. Human myoblast transplantation between immunohistocompatible donors and recipients produces immune reactions. Transplant Proc 1992; 24: 3049–51 NAN 2015; 41: 270–287

Advances in cell therapy strategies

118 Gussoni E, Pavlath GK, Lanctot AM, Sharma KR, Miller RG, Steinman L, Blau HM. Normal dystrophin transcripts detected in Duchenne muscular dystrophy patients after myoblast transplantation. Nature 1992; 356: 435–8 119 Karpati G, Ajdukovic D, Arnold D, Gledhill RB, Guttmann R, Holland P, Koch PA, Shoubridge E, Spence D, Vanasse M, Watters G, Abrahamowicz M, Duff C, Worton RG. Myoblast transfer in Duchenne muscular dystrophy. Ann Neurol 1993; 34: 8–17 120 Tremblay JP, Malouin F, Roy R, Huard J, Bouchard JP, Satoh A, Richards CL. Results of a triple blind clinical study of myoblast transplantations without immunosuppressive treatment in young boys with Duchenne muscular dystrophy. Cell Transplant 1993; 2: 99– 112 121 Tremblay JP, Bouchard JP, Malouin F, Theau D, Cottrell F, Collin H, Rouche A, Gilgenkrantz S, Abbadi N, Tremblay M, Tomé FMS, Fardeau M. Myoblast transplantation between monozygotic twin girl carriers of Duchenne muscular dystrophy. Neuromuscul Disord 1993; 3: 583–92 122 Morandi L, Bernasconi P, Gebbia M, Mora M, Crosti F, Mantegazza R, Cornelio F. Lack of mRNA and dystrophin expression in DMD patients three months after myoblast transfer. Neuromuscul Disord 1995; 5: 291–5 123 Mendell JR, Kissel JT, Amato AA, King W, Signore L, Prior TW, Sahenk Z, Benson S, McAndrew PE, Rice R, Nagaraja H, Stephens R, Lantry L, Morris GE, Burghes AHM. Myoblast transfer in the treatment of Duchenne’s muscular dystrophy. N Engl J Med 1995; 333: 832–8 124 Miller RG, Sharma KR, Pavlath GK, Gussoni E, Mynhier M, Lanctot AM, Greco CM, Steinman L, Blau HM. Myoblast implantation in Duchenne muscular dystrophy: the San Francisco study. Muscle Nerve 1997; 20: 469–78 125 Neumeyer AM, Cros D, McKenna-Yasek D, Zawadzka A, Hoffman EP, Pegoraro E, Hunter RG, Munsat TL, Brown RH Jr. Pilot study of myoblast transfer in the treatment of Becker muscular dystrophy. Neurology 1998; 51: 589–92 126 Skuk D, Roy B, Goulet M, Chapdelaine P, Bouchard JP, Roy R, Dugre FJ, Lachance JG, Deschenes L, Helene S, Sylvain M, Tremblay JP. Dystrophin expression in myofibers of Duchenne muscular dystrophy patients following intramuscular injections of normal myogenic cells. Mol Ther 2004; 9: 475–82 127 Torrente Y, Belicchi M, Marchesi C, Dantona G, Cogiamanian F, Pisati F, Gavina M, Giordano R, Tonlorenzi R, Fagiolari G, Lamperti C, Porretti L, Lopa R, Sampaolesi M, Vicentini L, Grimoldi N, Tiberio F, Songa

© 2014 British Neuropathological Society

128

129

130

131

132

133 134 135

136

137

287

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. Autologous transplantation of muscle-derived CD133+ stem cells in Duchenne muscle patients. Cell Transplant 2007; 16: 563–77 Perie S, Trollet C, Mouly V, Vanneaux V, Mamchaoui K, Bouazza B, Marolleau JP, Laforet P, Chapon F, Eymard B, Butler-Browne G, Larghero J, St Guily JL. Autologous myoblast transplantation for oculopharyngeal muscular dystrophy: a phase I/IIa clinical study. Mol Ther 2013; 22: 219–25 Sebe P, Doucet C, Cornu JN, Ciofu C, Costa P, de Medina SG, Pinset C, Haab F. Intrasphincteric injections of autologous muscular cells in women with refractory stress urinary incontinence: a prospective study. Int Urogynecol J 2011; 22: 183–9 Frudinger A, Kolle D, Schwaiger W, Pfeifer J, Paede J, Halligan S. Muscle-derived cell injection to treat anal incontinence due to obstetric trauma: pilot study with 1 year follow-up. Gut 2009; 59: 55–61 Blaganje M, Lukanovic A. Intrasphincteric autologous myoblast injections with electrical stimulation for stress urinary incontinence. Int J Gynaecol Obstet 2013; 117: 164–7 Cooper RN, Thiesson D, Furling D, Di Santo JP, Butler-Browne GS, Mouly V. Extended amplification in vitro and replicative senescence: key factors implicated in the success of human myoblast transplantation. Hum Gene Ther 2003; 14: 1169–79 Kipling D, Cooke HJ. Hypervariable ultra-long telomeres in mice. Nature 1990; 347: 400–2 Kipling D. Telomeres, replicative senescence and human ageing. Maturitas 2001; 38: 25–37 Trollet C, Gidaro T, Klein P, Perie S, Butler-Browne G, Lacau St Guily J. Oculopharyngeal muscular dystrophy. 1993 Coiffier L, Perie S, Laforet P, Eymard B, Lacau St Guily J. Long-term results of cricopharyngeal myotomy in oculopharyngeal muscular dystrophy. Otolaryngol Head Neck Surg 2006; 135: 218–22 Perie S, Mamchaoui K, Mouly V, Blot S, Bouazza B, Thornell LE, St Guily JL, Butler-Browne G. Premature proliferative arrest of cricopharyngeal myoblasts in oculo-pharyngeal muscular dystrophy: therapeutic perspectives of autologous myoblast transplantation. Neuromuscul Disord 2006; 16: 770–81

Received 1 July 2014 Accepted after revision 14 November 2014 Published online Article Accepted on 18 November 2014

NAN 2015; 41: 270–287

Suggest Documents