111
Views
46
CrossRef citations to date
0
Altmetric
Review

Myoblast transplantation for inherited myopathies: a clinical approach

Pages 1871-1885 | Published online: 22 Feb 2005

Bibliography

  • PARTRIDGE TA, GROUNDS M, SLOPER JC: Evidence of fusion between host and donor myoblasts in skeletal muscle grafts. Nature (1978) 273:306–308.
  • JEPPESEN J, GREEN A,STEFFENSEN BE RAHBEK J: The Duchenne muscular dystrophy population in Denmark, 1977-2001: prevalence, incidence and survival in relation to theintroduction of ventilator use. Neureinuscul. Diserd. (2003) 13:804–812.
  • LIPTON BH, SCHULTZ E: Developmental fate of skeletal muscle satellite cells. Science (1979) 205:1292–1294.
  • •This pioneer paper (one of the first two publications on MT) initially described that following intramuscular injection, donor myoblasts either fuse together to form new small myofibres or fuse with the recipient's myofibres to be incorporated in large myofibres.
  • 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. Neurel. Sci. (1982) 57:319–331.
  • •Another pioneer paper on MT, which demonstrated that the expression of donor and host proteins coexist in skeletal muscles of mice following MT.
  • PARTRIDGE TA, MORGAN JE,COULTON GR, HOFFMAN EP,KUNKEL LM: Conversion of mdxmyofibres from dystrophin-negative to-positive by injection of normal myoblasts. Nature (1989) 337:176–179.
  • ••A seminal paper establishing theprinciple of cell transplantation as a means of introducing dystrophin into dystrophic muscle. This paper also identified some of the main problems of myoblast transplantation.
  • SKUK D, ROY B, GOULET M et al.: Dystrophin expression in myofibers of Duchenne muscular dystrophy patients following intramuscular injections of normal myogenic cells. Ma The]: (2004) 9:475–482.
  • ••This report was the first demonstration inhumans that normal MT can systematically induce dystrophin expression in significant percentages of myofibres in DMD patients, if an appropriate donor cell delivery and immunosuppression are used.
  • MENDELL JR, KISSEL JT, AMATO AA et al.: Myoblast transfer in the treatment of Duchenne's muscular dystrophy. N Engl. J. Med. (1995) 333:832–838.
  • PAVLATH GK, RICH K, WEBSTER SG, BLAU HM: Localization of muscle gene products in nuclear domains. Nature (1989) 337:570–573.
  • •This study determined that proteins coded by a single nucleus in a myofibre are expressed throughout a limited region of the syncytium, which is called the 'nuclear domain'. This fmding implies that the expression of donor proteins in a myofibre, following the fusion of a donor myoblast, is restricted to a limited region and not to the entire sarcolemma.
  • RALSTON E, HALL ZW: Restricteddistribution of mRNA produced from a single nucleus in hybrid myotubes. J. Cell Biol. (1992) 119:1063–1068.
  • •A study that determined the short diffusion of the mRNA produced in the nucleus of a myofibre, which was estimated to be 1001.1m.
  • HALL ZW, RALSTON E: Nuclear domains in muscle cells. Cell (1989) 59:771–772.
  • KARPATI G, POULIOT Y, ZUBRZYCKA-GAARN E et al.: Dystrophin is expressed in mdx skeletal muscle fibers after normal myoblast implantation. Am. Pathol (1989) 135:27–32.
  • WERNIG A, IRINTCHEV A, LANGE G: Functional effects of myoblast implantation into histoincompatible mice with orwithout immunosuppression. I Physiol (Lond) (1995) 484:493–504.
  • •A detailed study on the potential of MT to restore muscular mass in mice following acute irreversible damage, with special interest in immunological aspects.
  • IRINTCHEV A, LANGER M,ZWEYER M, THEISEN R, WERNIG A: Functional improvement of damaged adult mouse muscle by implantation of primary myoblasts. Physiol. (Lond) (1997) 500:775–785.
  • •A study on the potential of MT to restore muscular mass in mice following acute irreversible damage.
  • ALAMEDDINE HS, LOUBOUTIN JP, DEHAUPAS M, SEBILLE A,FARDEAU M: Functional recovery induced by satellite cell grafts in irreversibly injured muscles. Cell Transplant. (1994) 3:3–14.
  • •One of the first studies on mice demonstrating that MT can restore mass and force in acutely damaged skeletal muscles in which endogenous regeneration was inhibited.
  • WERNIG A, ZWEYER M, IRINTCHEV A: Function of skeletal muscle tissue formed after myoblast transplantation into irradiated mouse muscles. J. Physic] (Lond) (2000) 522:333–345.
  • YAO SN, KURACHI K: Implanted myoblasts not only fuse with myofibers but also survive as muscle precursor cells. J. Cell Sci. (1993) 105:957–963.
  • •The first study to demonstrate that mice donor myoblasts can remain as dormant mononuclear muscle-precursor cells in the host muscle following MT. This fmding implies that, if the same could be obtained in humans, clinical MT could provide a source of normal myogenic cells able to participate later in hypertrophy and muscle regeneration.
  • GROSS JG, MORGAN JE: Muscleprecursor cells injected into irradiated mdxmouse muscle persist after serial injury. Muscle Nerve (1999) 22:174–185.
  • •This study provided further evidence that donor myoblasts can remain as mononuclear muscle-precursor cells in the host muscle following MT in mice.
  • HESLOP L, BEAUCHAMP JR,TAJBAKHSH S, BUCKINGHAM ME, PARTRIDGE TA, ZAMMIT PS: Transplanted primary neonatal myoblasts can give rise to functional satellite cells asidentified using the Myf5(nlacZ1+) mouse. Gene The]: (2001) 8:778–783.
  • ••This paper demonstrated for the first timethat at least some of the donor myoblasts, which remain as mononudeated precursor cells in host muscles, stay specifically in the form of satellite cells. If this is confirmed in humans, it implies that MT could not induce the expression ofnormal proteins in the myofibres of myopathic patients, but could restore a pool of functional satellite cells (at this time normal).
  • MAURO A: Satellite cell of skeletal musclefibers. J. Biophys. Biochetn. Cytol. (1961) 9:493–495.
  • •A landmark paper that reported the discovery of satellite cells.
  • LEWIS MR: Rhythmical contraction of the skeletal muscle tissue observed in tissue cultures. Am. J. Physic] (1915) 38:153–161.
  • KONIGSBERG IR: The differentiation of cross-striated myofibrils in short term cell culture. Exp. Cell Res. (1960) 21:414–420.
  • KINOSHITA I, ROY R, DUGRE FJ et al.: Myoblast transplantation in monkeys: control of immune response by FK506. Neuropathol Exp. Neurol (1996) 55:687–697.
  • •This was the first approach to the study of the acute rejection response following MT in non-human primates.
  • KINOSHITA I, VILQUIN JT,GRAVEL C, ROY R, TREMBLAY JP: Myoblast allotransplantation in primates [letter]. Muscle Nerve (1995) 18:1217–1218.
  • •This was the first preclinical study of MT in non-human primates, which demonstrated that hybrid myofibres can be obtained in this model following MT under tacrolimus immunosuppression.
  • SKUK D, ROY B, GOULET M, TREMBLAY JP: Successful myoblast transplantation in primates depends on appropriate cell delivery and induction of regeneration in the host muscle. Exp. Neurol (1999) 155:22–30.
  • •This paper initially demonstrated that it was possible to obtain a high performance of MT in the muscles of a primate, thus indicating no inherent difference between the results of mouse MT and human MT, if similar parameters are applied.
  • SKUK D, GOULET M, ROY B, TREMBLAY JP: Efficacy of myoblast transplantation in nonhuman primates following simple intramuscular cell injections: toward defining strategies applicable to humans. Exp. Neurol (2002) 175:112–126.
  • •This paper showed the possibility to reach high percentages of hybrid myofibres in the skeletal muscles of non-human primates following the intramuscular implantation of myoblasts, without myotoxins. It was the first paper to establish a quantitative parameter in order to make MT a technique to provide reproducible results in large muscles of primates.
  • SKUK D, GOULET M, ROY B, TREMBLAY JP: Myoblast transplantation in whole muscle of nonhuman primates. Neuropathol Exp. Neurol (2000) 59:197–206.
  • ••Through this study, it is shown that MTcan be successfully carried out throughout large-limb muscles in primates if a method of HDI is used. It also demonstrates that muscles tolerate this treatment well, in spite of the initial inflammatory reaction, and that muscle damage, if maintained under some limits, did not cause the systemic toxicity of a rhabdomyolysis. Finally, the beneficial properties of tacrolimus as an immunosuppressant to control acute rejection in MT, while preserving the success of the graft, were confirmed in experiments of up to 1 year.
  • DEASY BM, JANKOWSKI RJ, HUARD J: Muscle-derived stem cells: characterization and potential for cell- mediated therapy. Blood Cells MM. Dis. (2001) 27:924–933.
  • QU-PETERSEN Z, DEASY B, JANKOWSKI R et al.: Identification of a novel population of muscle stem cells in mice: potential for muscle regeneration. Cell Biol. (2002) 157:851–864.
  • MUELLER GM, O& DAY T,WATCHKO JF, ONTELL M: Effect of injecting primary myoblasts versus putative muscle-derived stem cells on mass and force generation in mdx mice. Hum. Gene Thec (2002) 13:1081–1090.
  • WERNIG A, IRINTCHEV A, HARTLING A, STEPHAN G, ZIMMERMANN K, STARZINSKI-POWITZ A: Formation of new muscle fibres and tumours after injection of cultured myogenic cells. Neurocytol (1991) 20:982–997.
  • FRESHNEY RI: Culture of Animal Cells. A Manual of Basic Technique. Freshney RI (Ed.), Alan R Liss, Inc., New York, USA (1987).
  • TREMBLAY JP, ROY B, GOULET M: Human myoblast transplantation: a simple assay for tumorigenicity. Neuromuscul Disord. (1991) 1:341–343.
  • GRIGORIADIS AE, HEERSCHE JN, AUBIN JE: Differentiation of muscle, fat, cartilage, and bone from progenitor cells present in a bone-derived clonal cell population: effect of dexamethasone. Cell Biol. (1988) 106:2139–2151.
  • WAKITANI S, SAITO T, CAPLAN Al: Myogenic cells derived from rat bone marrow mesenchymal stem cells exposed to 5-azacytidine. Muscle Nerve (1995) 18:1417–1426.
  • FERRARI G, CUSELLA-DE ANGELIS G, COLETTA M et al.: Muscle regeneration by bone marrow-derived myogenic progenitors. Science (1998) 279:1528–1530.
  • GUSSONI E, SONEOKA Y, STRICKLAND CD et al: Dystrophin expression in the mdx mouse restored by stem cell transplantation. Nature (1999) 401:390–394.
  • GUSSONI E, BENNETT RR, MUSKIEWICZ KR et al.: Long-term persistence of donor nuclei in a Duchenne muscular dystrophy patient receiving bone marrow transplantation. J. Gin. Invest. (2002) 110:807–814.
  • CAMARGO FD, GREEN R, CAPETENAKI Y, JACKSON KA, GOODELL MA: Single hematopoietic stem cells generate skeletal muscle through myeloid intermediates. Nat. Med. (2003) 9:1520–1527.
  • ••An interesting study that raised thepossibility that the rare fusion of circulating bone marrow-derived cells with myofibres should be carried out by the occasional recruitment of a macrophage in the fusiogenic process during myofibre regeneration. The rationality of this study comes from the fact that macrophages are cells that have the capacity to fuse, and that they densely infiltrate necrosed myofibres, participating actively in the process of muscle regeneration.
  • HUARD C, MOISSET PA, DICAIRE A et al.: Transplantation of dermal fibroblasts expressing MyoD1 in mouse muscles. Biochem. Biophys. Res. Commun. (1998) 248:648–654.
  • GOLDRING K, JONES GE, SEWRY CA, WATT DJ: The muscle-specific marker desmin is expressed in a proportion of human dermal fibroblasts after theirexposure to galectin-1. Neuromuscul Disord. (2002) 12:183–186.
  • LANDON DN: Skeletal muscle: normal morphology, development and innervation. In: Skeletal Muscle Pathology Mastaglia FL, Walton JN (Eds), Churchill Livingstone, Edinburgh, UK (1982):1–87.
  • PARTRIDGE TA: Invited review: myoblast transfer: a possible therapy for inherited myopathies? Muscle Nerve (1991) 14:197–212.
  • ••Probably the best review on MT duringthe beginning of this technique.
  • NEUMEYER AM, DIGREGORIO DM, BROWN RH JR: Arterial delivery of myoblasts to skeletal muscle. Neurology (1992) 42:2258–2262.
  • TORRENTE Y, TREMBLAY JP, PISATI F et al.: Intraarterial injection of muscle-derived CD34+Sca-1+ stem cells restores dystrophin in mdx mice. .1 Cell Biol. (2001) 152:335–348.
  • SAMPAOLESI M, TORRENTE Y, INNOCENZI A et al.: Cell therapy of alpha-sarcoglycan null dystrophic mice through intra-arterial delivery of mesoangioblasts. Science (2003) 301:487–492.
  • MILLER RG, SHARMA KR, PAVLATH GK et al.: Myoblast implantation in Duchenne muscular dystrophy: the San Francisco study. Muscle Nerve (1997) 20:469–478.
  • NEUMEYER AM, CROS D,MCKENNA-YASEK D et al.: Pilot study of myoblast transfer in the treatment of Becker muscular dystrophy. Neurology (1998) 51:589–592.
  • MORANDI L, BERNASCONI P, GEBBIA M et al.: Lack of mRNA and dystrophin expression in DMD patients three months after myoblast transfer. Neuromuscul Disord. (1995) 5:291–295.
  • TREMBLAY JP, MALOUIN F, ROY R et al.: 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.
  • TREMBLAY JP, BOUCHARD JP, MALOUIN F et al.: Myoblast transplantation between monozygotic twin girl carriers of Duchenne muscular dystrophy. Neuron/use& Disord. (1993) 3:583–592.
  • KARPATI G, JOHNSTON W, AJDUKOVIC G et al: Myoblast transfer (MT) in McArdle's disease (McD). Neurology (1992) 42\(Suppl. 3):387.
  • KARPATI G, AJDUKOVIC D, ARNOLD D et al.: Myoblast transfer in Duchenne muscular dystrophy. Ann. Neurol (1993) 34:8–17.
  • HUARD J, BOUCHARD JP, ROY R et al:Human myoblast transplantation: preliminary results of 4 cases. Muscle Nerve (1992) 15:550–560.
  • GUSSONI E, PAVLATH GK,LANCTOT AM et al.: Normal dystrophin transcripts detected in Duchenne muscular dystrophy patients after myoblast transplantation. Nature (1992) 356:435–438.
  • WARD MM: Factors predictive of acute renal failure in rhabdomyolysis. Arch. Intern. Med. (1988) 148:1553–1557.
  • EL FAHIME E, TORRENTE Y, CARON NJ, BRES OLIN MD, TREMBLAY JP: ha vivo migration of transplanted myoblasts requires matrix metalloproteinase activity. Exp. Cell Res. (2000) 258:279–287.
  • EL FAHIME E, MILLS P,LAFRENIERE JF, TORRENTE Y, TREMBLAY JP: The urokinase plasminogen activator: an interesting way to improve myoblast migration following their transplantation. Exp. Cell Res. (2002) 280:169–178.
  • ITO H, HALLAUER PL, HASTINGS KE,TREMBLAY JP: Prior culture with concanavalin A increases intramuscular migration of transplanted myoblast. Muscle Nerve (1998) 21:291–297.
  • CARON NJ, ASSELIN I, MOREL G, TREMBLAY JP: Increased myogenic potential and fusion of matrilysin-expressing myoblasts transplanted in mice. Cell Transplant. (1999) 8:465–476.
  • HUARD J, VERREAULT S, ROY R, TREMBLAY M, TREMBLAY JP: High efficiency of muscle regeneration after human myoblast clone transplantation in SCID mice. J. Gin. Invest. (1994) 93:586–599.
  • KINOSHITA I, VILQUIN JT, GUERETTE B, ASSELIN I, ROY R, TREMBLAY JP: Very efficient myoblast allotransplantation in mice under FK506 immunosuppression. Muscle Nerve (1994) 17:1407–1415.
  • •This paper reported the best results obtained with MT in dystrophic mice,attributed to the use of tacrolirnus for immunosuppression.
  • VILQUIN JT, ASSELIN I, GUERETTE B,KINOSHITA I, ROY R, TREMBLAY JP: Successful myoblast allotransplantation in mdx mice using rapamycin. Transplantation (1995) 59:422–426.
  • •The benefits of raparnycin as anirnmunosuppressant for MT in dystrophic mice are reported in this paper. This was the only MT study in which raparnycin was used.
  • SKUK D, FURLING D, BOUCHARD JP et al.: Transplantation of human myoblasts in SCID mice as a potential muscular model for myotonic dystrophy. J. Neuropathol Exp. Neurol (1999) 58:921–931.
  • PIN CL, MERRIFIELD PA: Developmental potential of rat L6 myoblasts in vivo following injection into regenerating muscles. Dev. Biol. (1997) 188:147–166.
  • CANTINI M, MASSIMINO ML, CATANI C, RIZZUTO R, BRINI M, CARRARO U: Gene transfer into satellite cell from regenerating muscle: bupivacaine allows beta-Gal transfection and expression in vitro and in vivo. In Vitro Cell. Dev. Biol. Amin. (1994) 30A:131–133.
  • 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) 111:2437–2449.
  • ••This study demonstrated that normalMT can improve the histology in the skeletal muscles of dystrophic mice, in which muscle degeneration was accelerated by pre-irradiation.
  • KINOSHITA I, VILQUIN JT, ASSELIN I, CHAMBERLAIN J, TREMBLAY JP: Transplantation of myoblasts from a transgenic mouse overexpressing dystrophin produced only a relatively small increase of dystrophin-positive membrane. Muscle Nerve (1998) 21:91–103.
  • ZVIBEL I, SMETS F, SORIANO H: Anoikis: roadblock to cell transplantation? Cell Transplant. (2002) 11:621–630.
  • BEAUCHAMP JR, MORGAN JE, PAGEL CN, PARTRIDGE TA: Dynamics of myoblast transplantation reveal a discrete minority of precursors with stem cell-like properties as the myogenic source. J. Cell Biol. (1999) 144:1113–1122.
  • ••This paper established a good method toanalyse early donor cell survival followingMT, discriminating donor cell death from donor cell differentiation. In fact, it was the first observation that both phenomena develop simultaneously during the first post-transplantation days in such a way that cell proliferation compensates cell death, therefore allowing good transplantation results.
  • 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) 62:951–967.
  • •Evidence of apoptosis as a mechanism of early donor cell death is shown for the first time in this paper.
  • GUERETTE B, SKUK D, CELESTIN F et al.: Prevention by anti-LFA-1 of acute myoblast death following transplantation. J. _lulu-ulna (1997) 159:2522–2531.
  • SAMMELS LM, BOSIO E, FRAGALL CT, GROUNDS MD, VAN ROOIJEN N, BEILHARZ MW: Innate inflammatory cells are not responsible for early death of donor myoblasts after myoblast transfer therapy. Transplantation (2004) 77:1790–1797.
  • COUSINS JC, WOODWARD KJ, GROSS JG, PARTRIDGE TA,MORGAN JE: Regeneration of skeletal muscle from transplanted immortalised myoblasts is oligoclonal. J. Cell Sci. (2004) 117:3259–3269.
  • JONES PH: Implantation of culturedregenerate muscle cells into adult rat muscle. Exp. Neurol (1979) 66:602–610.
  • •This pioneer paper was one of the first publications on MT, and it described for the first time the acute rejection in this model.
  • GUERETTE B, ASSELIN I, VILQUIN JT,ROY R, TREMBLAY JP: Lymphocyte infiltration following allo- and xenomyoblast transplantation in mdx mice. Muscle Nerve (1995) 18:39–51.
  • •This was one of the first characterisations of the specific immune cell reactions following myoblast allotransplantation in mice.
  • IRINTCHEV A, ZWEYER M, WERNIG A: Cellular and molecular reactions in mouse muscles after myoblast implantation. J. Neurocytol (1995) 24:319–331.
  • •This was one of the first characterisations of the specific immune cell reactions following myoblast allotransplantation in mice.
  • WERNIG A, IRINTCHEV A: 'Bystander'damage of host muscle caused by implantation of MHC- compatible myogenic cells. J. Neurol Sci. (1995) 130:190–196.
  • GUERETTE B, ROY R, TREMBLAY M et al: Increased granzyme B mRNA after alloincompatible myoblast transplantation. Transplantation (1995) 60:1011–1016.
  • GUERETTE B, TREMBLAY G, VILQUIN JT et al.: Increased interferon-gamma mRNA expression following alloincompatible myoblast transplantation is inhibited by FK506. Muscle Nerve (1996) 19:829–835.
  • CAMIRAND G, CARON NJ, ASSELIN I, TREMBLAY JP: Combined immunosuppression of mycophenolate mofetil and FK506 for myoblast transplantation in mdx mice. Transplantation (2001) 72:38–44.
  • HARDIMAN 0, SKLAR RM,BROWN RH JR: Direct effects of cyclosporin A and cyclophosphamide on differentiation of normal human myoblasts in culture. Neurology (1993) 43:1432–1434.
  • HONG F, LEE J, SONG JVV et al.: 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 from cyclosporin A-induced cytotoxicity. FASEB (2002) 16:1633–1635.
  • IRINTCHEV A, ZWEYER M,COOPER RN, BUTLER-BROWNE GS, WERNIG A: Contractile properties, structure and fiber phenotype of intact and regenerating slow-twitch muscles of mice treated with cyclosporin A. Cell Tissue Res. (2002) 308:143–156.
  • KINOSHITA I, VILQUIN JT,GUERETTE B et al.: Immunosuppression with FK 506 insures good success of myoblast transplantation in MDX mice. Transplant. Proc. (1994) 26:3518.
  • SEMSARIAN C, WU MJ, JU YK et al.: Skeletal muscle hypertrophy is mediated by a Ca2+-dependent calcineurin signalling pathway. Nature (1999) 400:576–581.
  • •This paper raised the possibility that tacrolimus-based immunosuppression could inhibit the mechanism of hypertrophy in skeletal muscles by describing how calcineurin plays a role in the mechanism of skeletal muscle hypertrophy.
  • VILQUIN JT, KINOSHITA I, ROY R, TREMBLAY JP: Cyclophosphamide immunosuppression does not permit successful myoblast allotransplantation in mouse. Neuromuscul Disord. (1995) 5:511–517.
  • VILQUIN JT, KINOSHITA I, ROY B et al.: Partial laminin alpha2 chain restoration in alpha2 chain-deficient dy/dy mouse by primary muscle cell culture transplantation. J. Cell Biol. (1996) 133:185–197.
  • •The first demonstration in mice of the potential of MT to treat congenital muscular dystrophy (another myopathy).
  • WATT DJ, MORGAN JE,PARTRIDGE TA: Long term survival of allografted muscle precursor cells following a limited period of treatment with cyclosporin A. Clin. Exp. Immunol (1984) 55:419–426.
  • BRAZELTON TR, MORRIS RE: Molecular mechanisms of action of new xenobiotic immunosuppressive drugs: tacrolimus (FK506), sirolimus (rapamycin), mycophenolate mofetil and leflunomide. Curr. Opin. Immunol (1996) 8:710–720.
  • GUMMERT JF, IKONEN T,MORRIS RE: Newer immunosuppressive drugs: a review. J. Am. Soc. Nephrol (1999) 10:1366–1380.
  • PAVLATH GK, RANDO TA, BLAU HM: Transient immunosuppressive treatment leads to long-term retention of allogeneic myoblasts in hybrid myofibers. J. Cell Biol. (1994) 127:1923–1932.
  • CAMIRAND G, CARON NJ, TURGEON NA, ROSSINI AA, TREMBLAY JP: Treatment with anti-CD154 antibody and donor-specific transfusion prevents acute rejection of myoblast transplantation. Transplantation (2002) 73:453–461.
  • CAMIRAND G, ROUSSEAU J, DUCHARME ME, ROTHSTEIN DM, TREMBLAY JP: Novel Duchenne muscular dystrophy treatment through myoblast transplantation tolerance with anti-CD45RB, anti-CD154 and mixed chimerism. Am. J. Transplant. (2004) 4:1255–1265.
  • GILL RG, WOLF L: Immunobiology of cellular transplantation. Cell Transplant. (1995) 4:361–370.
  • WIENDL H, MITSDOERFFER M, HOFMEISTER V et al.: The non-classical MHC molecule HLA-G protects human muscle cells from immune-mediated lysis:implications for myoblast transplantation and gene therapy. Brain (2003) 126:176-185.An interesting in vitro study that proposed an innovative method to protect the transplanted myoblasts from acute rejection, by way of expressing HLA-G, a non-classical MHC class I molecule, which is a key mediator in maternal tolerance to the fetus.
  • FLOYD SS JR, CLEMENS PR,ONTELL MR et al.: Ex vivo gene transferusing adenovirus-mediated full-length dystrophin delivery to dystrophic muscles. Gene Ther. (1998) 5:19–30.
  • BUJOLD M, CARON N, CAMIRAN G et al.: Autotransplantation in mdx mice of mdx myoblasts genetically corrected by anHSV-1 amplicon vector. Cell Transplant. (2002) 11:759–767.
  • MOISSET PA, SKUK D, ASSELIN I et al: Successful transplantation of genetically corrected DMD myoblasts following ex vivo transduction with the dystrophin minigene. Biochem. Biophys. Res. Commun. (1998) 247:94–99.
  • •This paper showed that it is possible to genetically correct human DMD myoblasts in vitro, being grafted back (in this case in SCID mice) to form human myofibres expressing dystrophin.
  • MOISSET PA, GAGNON Y, KARPATI G, TREMBLAY JP: Expression of human dystrophin following the transplantation of genetically modified mdx myoblasts. Gene Ther. (1998) 5:1340–1346.
  • VILQUIN JT, WAGNER E,KINOSHITA I, ROY R, TREMBLAY JP:Successful histocompatible myoblast transplantation in dystrophin- deficient mdx mouse despite the production of antibodies against dystrophin. J. Cell Biol. (1995) 131:975–988.
  • •This was the first study on the potential immunogenicity of dystrophin alone, in the case of being expressed in dystrophin-deficient recipients.
  • OHTSUKA Y, UDAKA K, YAMASHIRO Y, YAGITA H, OKUMURA K: Dystrophin acts as a transplantation rejection antigen in dystrophin-deficient mice: implication for gene therapy. bronunol. (1998) 160:4635–4640.
  • •This study defined some epitopes of dystrophin that could be the most antigenic in mice, raising the possibility that they led to a slow T lymphocyte reaction.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.