Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
J Vis Exp ; (83): e50532, 2014 Jan 20.
Article in English | MEDLINE | ID: mdl-24472871

ABSTRACT

Patient-derived iPSCs could be an invaluable source of cells for future autologous cell therapy protocols. iPSC-derived myogenic stem/progenitor cells similar to pericyte-derived mesoangioblasts (iPSC-derived mesoangioblast-like stem/progenitor cells: IDEMs) can be established from iPSCs generated from patients affected by different forms of muscular dystrophy. Patient-specific IDEMs can be genetically corrected with different strategies (e.g. lentiviral vectors, human artificial chromosomes) and enhanced in their myogenic differentiation potential upon overexpression of the myogenesis regulator MyoD. This myogenic potential is then assessed in vitro with specific differentiation assays and analyzed by immunofluorescence. The regenerative potential of IDEMs is further evaluated in vivo, upon intramuscular and intra-arterial transplantation in two representative mouse models displaying acute and chronic muscle regeneration. The contribution of IDEMs to the host skeletal muscle is then confirmed by different functional tests in transplanted mice. In particular, the amelioration of the motor capacity of the animals is studied with treadmill tests. Cell engraftment and differentiation are then assessed by a number of histological and immunofluorescence assays on transplanted muscles. Overall, this paper describes the assays and tools currently utilized to evaluate the differentiation capacity of IDEMs, focusing on the transplantation methods and subsequent outcome measures to analyze the efficacy of cell transplantation.


Subject(s)
Induced Pluripotent Stem Cells/transplantation , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/physiology , Regeneration/physiology , Regenerative Medicine/methods , Animals , Disease Models, Animal , Induced Pluripotent Stem Cells/cytology , Mice , Models, Animal , Muscle Fibers, Skeletal/cytology , Muscle, Skeletal/cytology , Muscular Dystrophy, Animal/surgery
2.
PLoS One ; 8(5): e63016, 2013.
Article in English | MEDLINE | ID: mdl-23671652

ABSTRACT

Muscle satellite cells (SCs) are stem cells that reside in skeletal muscles and contribute to regeneration upon muscle injury. SCs arise from skeletal muscle progenitors expressing transcription factors Pax3 and/or Pax7 during embryogenesis in mice. However, it is unclear whether these fetal progenitors possess regenerative ability when transplanted in adult muscle. Here we address this question by investigating whether fetal skeletal muscle progenitors (FMPs) isolated from Pax3(GFP/+) embryos have the capacity to regenerate muscle after engraftment into Dystrophin-deficient mice, a model of Duchenne muscular dystrophy. The capacity of FMPs to engraft and enter the myogenic program in regenerating muscle was compared with that of SCs derived from adult Pax3(GFP/+) mice. Transplanted FMPs contributed to the reconstitution of damaged myofibers in Dystrophin-deficient mice. However, despite FMPs and SCs having similar myogenic ability in culture, the regenerative ability of FMPs was less than that of SCs in vivo. FMPs that had activated MyoD engrafted more efficiently to regenerate myofibers than MyoD-negative FMPs. Transcriptome and surface marker analyses of these cells suggest the importance of myogenic priming for the efficient myogenic engraftment. Our findings suggest the regenerative capability of FMPs in the context of muscle repair and cell therapy for degenerative muscle disease.


Subject(s)
Dystrophin/deficiency , Muscle, Skeletal/cytology , Muscular Dystrophy, Duchenne/surgery , Myoblasts, Skeletal/transplantation , Stem Cell Transplantation/methods , Animals , Cells, Cultured , Dystrophin/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Injections, Intramuscular , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/embryology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Animal/surgery , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/physiopathology , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts, Skeletal/metabolism , Myofibrils/genetics , Myofibrils/physiology , Myogenin/genetics , Myogenin/metabolism , PAX3 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Regeneration/physiology , Reverse Transcriptase Polymerase Chain Reaction , Satellite Cells, Skeletal Muscle/transplantation , Transcriptome
3.
J Vis Exp ; (73)2013 Mar 27.
Article in English | MEDLINE | ID: mdl-23568004

ABSTRACT

Duchenne muscular dystrophy (DMD) is a severe genetic neuromuscular disorder that affects 1 in 3,500 boys, and is characterized by progressive muscle degeneration. In patients, the ability of resident muscle satellite cells (SCs) to regenerate damaged myofibers becomes increasingly inefficient. Therefore, transplantation of muscle progenitor cells (MPCs)/myoblasts from healthy subjects is a promising therapeutic approach to DMD. A major limitation to the use of stem cell therapy, however, is a lack of reliable imaging technologies for long-term monitoring of implanted cells, and for evaluating its effectiveness. Here, we describe a non-invasive, real-time approach to evaluate the success of myoblast transplantation. This method takes advantage of a unified fusion reporter gene composed of genes (firefly luciferase [fluc], monomeric red fluorescent protein [mrfp] and sr39 thymidine kinase [sr39tk]) whose expression can be imaged with different imaging modalities. A variety of imaging modalities, including positron emission tomography (PET), single-photon emission computed tomography (SPECT), magnetic resonance imaging (MRI), optical imaging, and high frequency 3D-ultrasound are now available, each with unique advantages and limitations. Bioluminescence imaging (BLI) studies, for example, have the advantage of being relatively low cost and high-throughput. It is for this reason that, in this study, we make use of the firefly luciferase (fluc) reporter gene sequence contained within the fusion gene and bioluminescence imaging (BLI) for the short-term localization of viable C2C12 myoblasts following implantation into a mouse model of DMD (muscular dystrophy on the X chromosome [mdx] mouse). Importantly, BLI provides us with a means to examine the kinetics of labeled MPCs post-implantation, and will be useful to track cells repeatedly over time and following migration. Our reporter gene approach further allows us to merge multiple imaging modalities in a single living subject; given the tomographic nature, fine spatial resolution and ability to scale up to larger animals and humans, PET will form the basis of future work that we suggest may facilitate rapid translation of methods developed in cells to preclinical models and to clinical applications.


Subject(s)
Luminescent Measurements/methods , Molecular Imaging/methods , Myoblasts/cytology , Myoblasts/transplantation , Stem Cell Transplantation/methods , Animals , Genes, Reporter , Mice , Muscular Dystrophy, Animal/surgery , Muscular Dystrophy, Duchenne/surgery , Positron-Emission Tomography , Transfection
4.
Exp Neurol ; 220(1): 212-6, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19682990

ABSTRACT

Muscular dystrophies (MDs) consist of a genetically heterogeneous group of disorders, recessive or dominant, characterized by progressive skeletal muscle weakening. To date, no effective treatment is available. Experimental strategies pursuing muscle regeneration through the transplantation of stem cell preparations have brought hope to patients affected by this disorder. Efficacy has been demonstrated in recessive MD models through contribution of wild-type nuclei to the muscle fiber heterokaryon; however, to date, there has been no study investigating the efficacy of a cell therapy in a dominant model of MD. We have recently demonstrated that Pax3-induced embryonic stem (ES) cell-derived myogenic progenitors are able to engraft and improve muscle function in mdx mice, a recessive mouse model for Duchenne MD. To assess whether this therapeutic effect can be extended to a dominant type of muscle disorder, here we transplanted these cells into FRG1 transgenic mice, a dominant model that has been associated with facioscapulohumeral muscular dystrophy. Our results show that Pax3-induced ES-derived myogenic progenitors are capable of significant engraftment after intramuscular or systemic transplantation into Frg1 mice. Analyses of contractile parameters revealed functional improvement in treated muscles of male mice, but not females, which are less severely affected. This study is the first to use Frg1 transgenic mice to assess muscle regeneration as well as to support the use of a cell-based therapy for autosomal dominant types of MD.


Subject(s)
Muscle, Skeletal/surgery , Muscular Dystrophy, Animal/surgery , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/transplantation , Stem Cell Transplantation/methods , Stem Cells/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Disease Models, Animal , Female , Genes, Dominant/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/therapeutic use , Male , Mice , Mice, Transgenic , Microfilament Proteins , Muscle Development/genetics , Muscle Weakness/genetics , Muscle Weakness/metabolism , Muscle Weakness/surgery , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Nuclear Proteins/genetics , PAX3 Transcription Factor , Paired Box Transcription Factors/metabolism , Paired Box Transcription Factors/pharmacology , Paired Box Transcription Factors/therapeutic use , RNA-Binding Proteins , Regeneration/genetics , Satellite Cells, Skeletal Muscle/cytology , Sex Characteristics , Stem Cells/cytology , Stem Cells/drug effects , Treatment Outcome
5.
Stem Cells ; 27(10): 2478-87, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19575422

ABSTRACT

Changes that occur in the skeletal muscle environment with the progress of muscular dystrophies may affect stem cell function and result in impaired muscle regeneration. It has previously been suggested that the success of stem cell transplantation could therefore be dependent both on the properties of the cell itself and on the host muscle environment. Here we engrafted young and mature adult mdx-nude mice, which are the genetic homolog of Duchenne muscular dystrophy, with a small number of satellite cells freshly isolated from young, normal donor mice. We found that the donor satellite cells contributed to muscle regeneration and self-renewal as efficiently within mature adult, as in young, dystrophic host muscle. Donor-derived satellite cells also contributed to robust regeneration after further injury, showing that they were functional despite the more advanced dystrophic muscle environment. These findings provide evidence that muscle tissue in a later stage of dystrophy may be effectively treated by stem cells.


Subject(s)
Graft Survival/physiology , Muscular Dystrophy, Animal/surgery , Regeneration/physiology , Satellite Cells, Skeletal Muscle/physiology , Satellite Cells, Skeletal Muscle/transplantation , Stem Cell Transplantation/methods , Age Factors , Animals , Cell Communication/physiology , Cell Survival/physiology , Disease Models, Animal , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Satellite Cells, Skeletal Muscle/cytology , Treatment Outcome
6.
Transplantation ; 87(9): 1275-82, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19424025

ABSTRACT

BACKGROUND: The use of nonautologous stem cells isolated from healthy donors for stem-cell therapy is an attractive approach, because the stem cells can be culture expanded in advance, thoroughly tested, and formulated into off-the-shelf medicine. However, human leukocyte antigen compatibility and related immunosuppressive protocols can compromise therapeutic efficacy and cause unwanted side effects. METHODS: Mesenchymal stem cells (MSCs) have been postulated to possess unique immune regulatory function. We explored the immunomodulatory property of human and porcine MSCs for the treatment of delta-sarcoglycan-deficient dystrophic hamster muscle without immunosuppression. Circulating and tissue markers of inflammation were analyzed. Muscle regeneration and stem-cell fate were characterized. RESULTS: Total white blood cell counts and leukocyte-distribution profiles were similar among the saline- and MSC-injected dystrophic hamsters 1 month posttreatment. Circulating levels of immunoglobulin A, vascular cell adhesion molecule-1, myeloperoxidase, and major cytokines involved in inflammatory response were not elevated by MSCs, nor were expression of the leukocyte common antigen CD45 and the cytokine transcriptional activator NF-kappaB in the injected muscle. Treated muscles exhibited increased cell-cycle activity and attenuated oxidative stress. Injected MSCs were found to be trapped in the musculature, contribute to both preexisting and new muscle fibers, and mediates capillary formation. CONCLUSIONS: Intramuscular injection of nonautologous MSCs can be safely used for the treatment of dystrophic muscle in immunocompetent hosts without inflaming the host immune system.


Subject(s)
Bone Marrow Cells/cytology , Mesenchymal Stem Cell Transplantation/methods , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/surgery , Transplantation, Heterologous/methods , Animals , Bone Marrow Transplantation , Cricetinae , Humans , Immunosuppressive Agents/therapeutic use , Injections, Intramuscular , Lipid Peroxidation , Male , Muscular Dystrophy, Animal/immunology , Regeneration , Reverse Transcriptase Polymerase Chain Reaction , Sarcoglycans/deficiency , Tissue Donors
7.
Transplantation ; 85(12): 1791-801, 2008 Jun 27.
Article in English | MEDLINE | ID: mdl-18580473

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy is a fatal genetic disease caused by lack of dystrophin. Myogenic cell transplantation (MT), a potential therapy for Duchenne muscular dystrophy, can restore dystrophin expression in muscles. Because allogeneic MT is highly resistant to peripheral tolerance, we proposed to induce central tolerance. However, given its immunogenicity, we asked whether central tolerance to donor major histocompatibility complex would allow long-term expression of dystrophin, a tissue-specific neoantigen in dystrophic recipients. METHODS: Central tolerance was induced in C57BL/10J mdx (dystrophic) mice by allogeneic bone marrow transplantation (BMT) after conditioning with either lethal total body irradiation (TBI) or an established nonmyeloablative protocol (anti-CD154, anti-CD8 mAbs, and low-dose TBI). Recipients subsequently received donor-strain MT or skin grafts. RESULTS: Long-term hematopoietic chimeras generated using either lethal TBI or the nonmyeloablative regimen were tolerant to donor skin grafts and both primary and secondary donor MT (>90 days). Myogenic cell transplantation survival was decreased when chimerism was transient, which was most common with nonmyeloablative conditioning and fully rather than haplo-mismatched donors. Interestingly, regardless of conditioning, MT was associated with localized muscle infiltration with Foxp3CD4, CD25CD4, and PerforinCD8 cells, whereas skin grafts lacked infiltration. CONCLUSIONS: Central tolerance achieved using regimens that eliminate nearly all endogenous peripheral lymphocytes (i.e., lethal irradiation) or a nonmyeloablative protocol that depleted peripheral CD8 cells, results in lymphocytic infiltration in muscles that received MT but not in skin allografts. This suggests that muscle-specific infiltration may result from lack of negative selection for peripheral neoantigens in the thymus after BMT and that tolerance after MT may rely on peripheral regulatory mechanisms.


Subject(s)
Antigens/immunology , Cell Transplantation , Dystrophin/immunology , Immune Tolerance/immunology , Muscle, Skeletal/cytology , Muscle, Skeletal/immunology , Animals , Bone Marrow Transplantation/immunology , CD4-Positive T-Lymphocytes/pathology , CD40 Ligand/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/pathology , Chimera/immunology , Disease Models, Animal , Dystrophin/metabolism , Immune Tolerance/drug effects , Immune Tolerance/radiation effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/immunology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/surgery , Skin Transplantation/immunology , Whole-Body Irradiation
8.
Nature ; 450(7173): E23; discussion E23-5, 2007 Dec 20.
Article in English | MEDLINE | ID: mdl-18097347

ABSTRACT

Human muscular dystrophies are devastating and incurable inherited diseases. Hopes of progress towards therapy of muscular dystrophies were aroused when Sampaolesi et al. reported "extensive recovery of dystrophin expression, normal muscle...function", and "remarkable clinical amelioration" in golden retriever muscular dystrophy dogs treated with 'mesoangioblasts'. Here I re-examine their results, showing how their assessments might be flawed and their conclusions overstated. Further studies will be required to evaluate fully the clinical potential of this work.


Subject(s)
Dog Diseases/surgery , Muscular Dystrophy, Animal/surgery , Stem Cell Transplantation , Aging , Animals , Bias , Controlled Clinical Trials as Topic/trends , Dog Diseases/genetics , Dog Diseases/physiopathology , Dogs , Dystrophin/genetics , Gene Expression Regulation , Humans , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/physiopathology , Reproducibility of Results
10.
Di Yi Jun Yi Da Xue Xue Bao ; 25(5): 498-502, 2005 May.
Article in Chinese | MEDLINE | ID: mdl-15897118

ABSTRACT

OBJECTIVE: To investigate the feasibility of using human bone marrow-derived mesenchymal stem cells (hBM- MSCs) for repairing the skeletal muscle sarcolemma lesions in mdx mice and characterize the distribution of the transplanted hBM-MSCs. METHODS: Eighteen 8- to 10-week-old immunosuppressed mdx mice received transplantation with 1x10(7) of hBM-MSCs (the fifth passage) with 3H-thymidine (3H-TdR) labeling by injection of the cells into the tail vein. The mice were killed at 24 h, 48 h, 2 weeks, and 1, 2 and 4 months after the transplantation, respectively, to measure the radioactivity in the tissues and organs. Dystrophin expression on the sarcolemma was detected by immunofluorescence analysis. RESULTS: One month after transplantation, the mice with cell transplantation showed greater radioactivity in most of the tissues and organs than the control mice, especially in the bone marrow, liver and spleen. The radioactivity was then gradually lowered but in the skeletal muscle, the radioactivity increased progressively since 2 weeks after transplantation, reaching the peak of 27.65+/-3.53 Bq/mg at 1 month. Compared with that in the control mice, the radioactivity in the bone marrow and skeletal muscle was persistently higher in mice with cell transplantation 1 month after transplantation. No dystrophin-positive cells were found in the mdx mice at 2 weeks but detected at 1 month. The percentage of dystrophin-positive fibers in each section ranged from a 6.6% (1 month) to 8.9% (4 months). CONCLUSIONS: hBM-MSCs engrafted in immunosuppressed mdx mice may differentiate into skeletal muscle cells to repair the pathological lesion of the skeletal muscle sarcolemma. The hBM-MSCs reside mainly in the bone marrow, liver and spleen in the early stage following transplantation, homing into the bone marrow and skeletal muscle later.


Subject(s)
Mesenchymal Stem Cell Transplantation , Muscular Dystrophy, Animal/surgery , Transplantation, Heterologous , Animals , Bone Marrow Cells/cytology , Dystrophin/metabolism , Female , Humans , Immunocompromised Host , Male , Mice , Mice, Inbred mdx , Sarcolemma , Staining and Labeling , Thymidine/analysis , Tritium
11.
Transplantation ; 78(8): 1172-6, 2004 Oct 27.
Article in English | MEDLINE | ID: mdl-15502715

ABSTRACT

BACKGROUND: Myoblast transfer therapy (MTT) is a strategy designed to compensate for the defective gene in myopathies such as Duchenne muscular dystrophy (DMD). Experimental MTT in the mdx mouse (an animal model of DMD) has used donor myoblasts derived from mice of various ages; however, to date, there has been no direct quantitative comparison between the efficacy of MTT using myoblasts isolated from adult and neonate donor muscle. METHODS: Donor normal male myoblasts were injected into Tibialis Anterior muscles of dystrophic female host mice and the survival and proliferation of male myoblasts quantitated using Y-chromosome specific real-time quantitative polymerase chain reaction. The survival of late preplate (PP6) myoblasts derived from neonatal (3-5 days old) or adult (6-8 weeks old) donor mice after MTT were compared. The influence of the number of tissue culture passages, on survival post-MTT, was also evaluated for both types of myoblasts. RESULTS: Surprisingly, superior transplantation efficiency was observed for adult-derived compared with neonatal myoblasts (both early and late passage). Extended expansion (>17 passages) in tissue culture resulted in inferior survival and proliferation of both adult and neonatal myoblasts; however, proliferation of early passage myoblasts (both adult and neonate) was evident between 3 weeks and 3 months. CONCLUSIONS: Myoblasts derived from neonatal mice were inferior for transplantation, and early passage donor myoblasts from adult mice are recommended for MTT in this model.


Subject(s)
Aging , Muscular Dystrophy, Animal/surgery , Myoblasts/transplantation , Animals , Animals, Newborn , Cell Count , Cell Division , Cell Survival , Cell Transplantation/methods , Cells, Cultured , Female , Injections, Intramuscular , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/surgery , Myoblasts/pathology , Time Factors , Tissue Donors
12.
Neuromuscul Disord ; 12 Suppl 1: S7-10, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12206789

ABSTRACT

Differentiated muscle fibres can be formed by transplanted haematopoietic stem cells in models of acute or chronic muscle regeneration, including the dystrophin-deficient mdx mouse. Muscle-forming activity can be found in adult, foetal and embryonic haematopoietic tissues. The blood-to-muscle transition may be due to transdifferentiation of haematopoietic progenitors in response to local signals provided by the regenerating muscle. These signals are only poorly provided by the muscle of the mdx mouse, since transplantation into these mice of normal C57Bl/6 bone marrow gives rise only to a minimal number of muscle fibres expressing the normal dystrophin protein (<1%) throughout the animal life span. Expansion and active recruitment to myogenic differentiation of transplanted haematopoietic cells are therefore critical factors for a future use of bone marrow transplantation in cell/gene therapy of muscular dystrophy.


Subject(s)
Bone Marrow Transplantation , Muscle Development , Muscular Dystrophies/surgery , Myoblasts/transplantation , Stem Cell Transplantation/methods , Animals , Dystrophin/deficiency , Genetic Therapy/trends , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscular Dystrophies/metabolism , Muscular Dystrophy, Animal/surgery , Muscular Dystrophy, Duchenne/surgery
13.
Muscle Nerve ; 23(4): 560-74, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10716768

ABSTRACT

Sliced male C57Bl/10Sn (H2-b) donor muscles were grafted into the female histocompatible muscles of untreated, FK506-treated, and T-cell depleted (with or without thymic tolerization) dystrophic (mdx; H2-b) and normal (C57Bl/10Sn; H2-b) hosts, and also into histoincompatible normal (Balb/c; H2-d) hosts. The fate of male donor nuclei was monitored on tissue sections by in situ hybridization with a Y-chromosome specific probe. The results demonstrate that the dystrophic environment is more conducive than normal muscle to donor myoblast migration, with the distance moved being threefold greater at 12 weeks in dystrophic hosts. T-cell depletion was significantly more effective than FK506 treatment at enhancing donor myoblast emigration in both histocompatible and histoincompatible hosts at 3 weeks. Furthermore, the effects of T-cell depletion were sustained in histoincompatible hosts at 12 weeks. These data endorse the use of host T-cell depletion as a promising long-term strategy to improve myoblast transfer therapy (MTT) in the clinical situation.


Subject(s)
Muscle, Skeletal/physiology , Muscle, Skeletal/transplantation , Muscular Dystrophy, Animal/surgery , Animals , Cell Movement , Female , Immunosuppression Therapy/methods , Lymphocyte Depletion , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/cytology , T-Lymphocytes/immunology , Tacrolimus/therapeutic use , Transplantation Chimera , Transplantation, Homologous , Y Chromosome
14.
Neuromuscul Disord ; 8(2): 95-110, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9608563

ABSTRACT

Dog myoblasts obtained from muscle biopsies were infected in vitro with a defective retroviral vector containing a cytoplasmic beta-galactosidase (beta-Gal) gene. These myoblasts were initially transplanted in the irradiated muscles of SCID mice and beta-Gal positive muscle fibers were observed. beta-Gal myoblasts were also transplanted back either in the donor dogs (autotransplantation model) or in unrelated recipient dogs (allotransplantation model). Following these myoblast injections, a rapid inflammatory reaction developed within the muscle as indicated by an expression of P-selectin and of pro-inflammatory cytokine mRNAs (interleukin 6 (IL-6) and transforming growth factor beta (TGF-beta), and by a neutrophil infiltration. Following either auto- or allotransplantation in inadequately or non-immunosuppressed dogs, a specific immune reaction also developed within 2 weeks as indicated by the infiltration of CD4+ and of CD8+ lymphocytes, the increased expression of IL-10 and granzyme B mRNAs and the presence of antibodies reacting with the injected cells. Some dogs were immunosuppressed with several combinations of FK506, cyclosporine (CsA) and RS-61443. In dogs immunosuppressed with CsA combined with RS-61443, only a few myoblasts and myotubes expressing beta-Gal were observed 1-2 weeks after the transplantation, but no muscle fibers expressing beta-Gal were observed after 4 weeks, and antibodies against the injected cells were formed. In dogs immunosuppressed with FK506 alone, although no antibodies against the injected cells were produced, there were no small cells and no muscle fibers expressing beta-Gal 1 month after the transplantation. However, FK506 triggered diarrhea and vomiting in dogs. When the dogs were immunosuppressed with FK506 combined with CsA and RS-61443, muscle fibers expressing beta-Gal were present 4 weeks after the transplantation and no antibodies reacting with donor myoblasts were detected. These results indicate that the combination of three immunosuppressive agents (i.e., FK506, CsA and RS-61443) is effective in controlling the specific immune reactions following myoblast transplantation in dogs and they underline that the outcome of myoblast transplantation is dependent in part on an adequate immunosuppression. These results obtained here in normal dogs may justify myoblast transplantation in dystrophic dogs despite the side effects of FK506.


Subject(s)
Cell Transplantation , Immunosuppressive Agents/therapeutic use , Muscles/transplantation , Animals , Biopsy , Cells, Cultured , Cyclosporine/therapeutic use , Dogs , Drug Therapy, Combination , Mice , Mice, SCID , Muscles/cytology , Muscles/immunology , Muscular Dystrophy, Animal/surgery , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/therapeutic use , Tacrolimus/therapeutic use , Transplantation, Heterologous , Transplantation, Homologous , beta-Galactosidase/biosynthesis
16.
Cell Transplant ; 5(3): 421-9, 1996.
Article in English | MEDLINE | ID: mdl-8727011

ABSTRACT

Excellent long-term survival (up to 1 yr) of donor skeletal muscle cells was demonstrated using a mouse Y-chromosome specific probe, following the transplantation of grafts of whole muscles from male "normal" C57B1/10Sn mice into dystrophic muscles of female host mice. After the transplantation of equivalent sliced muscle grafts there was extensive movement of the male donor cells and fusion with host myofibres. This contrasts with the extremely poor survival of isolated myoblasts after injection into the same mouse model for Duchenne muscular dystrophy. The use of sliced muscle grafts may therefore represent a potential alternative approach to myoblast transfer therapy.


Subject(s)
Cell Transplantation/methods , Muscle, Skeletal/transplantation , Muscular Dystrophy, Animal/surgery , Y Chromosome , Animals , Cell Movement , Dissection , Female , Graft Survival , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Transplantation, Homologous
18.
J Cell Biol ; 131(4): 975-88, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7490298

ABSTRACT

Myoblast transplantation has been considered a potential treatment for some muscular disorders. It has proven very successful, however, only in immunodeficient or immunosuppressed mice. In this study, myoblasts from C57BL10J +/+ mice were transplanted, with no immunosuppressive treatment, in the tibialis anterior of fully histocompatible but dystrophin-deficient C57BL10J mdx/mdx mice. One to 9 months after transplantation, the success of the graft was evaluated by immunohistochemistry. All the transplanted mice (n = 24) developed dystrophin-positive fibers following transplantation. Depending on myoblast cultures, transplantations, and time of analysis, the mice presented 15 to 80% of dystrophin-positive fibers in transplanted muscles. These fibers were correctly oriented and they were either from donor or hybrid origin. The dystrophin-positive fibers remained stable up to 9 months. Possible humoral and cellular immune responses were investigated after grafting. Antibodies directed against dystrophin and/or muscle membrane were developed by 58% of the mice as demonstrated by immunohistochemistry and Western blotting. Despite the presence of these antibodies, dystrophin-positive fibers were still present in grafted muscles 9 months after transplantation. Moreover, the muscles did not show massive infiltration by CD4 cells, CD8 cells, or macrophages, as already described in myoblast allotransplantations. This lack of rejection was attributed to the sequestrated nature of dystrophin after fiber formation. These results indicate that myoblast transplantation leads to fiber formation when immunocompetent but fully histocompatible donors and recipients are used and that dystrophin incompatibility alone is not sufficient to induce an immunological rejection reaction.


Subject(s)
Cell Transplantation , Dystrophin/deficiency , Dystrophin/immunology , Muscle, Skeletal/cytology , Muscular Dystrophy, Animal/surgery , Animals , Antibody Formation , Cell Movement/immunology , Disease Models, Animal , Dystrophin/genetics , Histocompatibility , Histocytochemistry , Immunoblotting , Lymphocytes/cytology , Mice , Mice, Inbred mdx , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/immunology , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Animal/immunology
19.
Gaoxiong Yi Xue Ke Xue Za Zhi ; 11(7): 398-408, 1995 Jul.
Article in Chinese | MEDLINE | ID: mdl-7650779

ABSTRACT

In this report, we study the suitable conditions for myoblast cultures through analysis of myoblast growth and differentiation, and then try to develop a mouse model for myoblast transfer therapy (MTT). Recently, some research has indicated that Muscular Dystrophy Murine Mice (MDX) have an X-linked recessive dystrophin deficiency which is caused by dystrophin gene point mutation at the X chromosome. Therefore, MDX mice are usually used for MTT models of muscular dystrophy disease. Control mice, C57BL10/SCSN (B-10) were chosen as a source of normal myoblasts. Myoblasts isolated from the hindlimb muscle tissues of two- to three-day-old neonatal B-10 mice were cultured in vitro for one to seven days. Through our modifyied techniques of isolation and culturing conditions, a myoblast purity of 70% could be achieved, with fibroblast the only contaminating cell type. The proliferative capacity and the doubling time of myoblasts were counted from analysis of growth kinetics. While differentiative capacity was analyzed morphologically, we found the fusion of myoblasts was time-dependent. Immunostaining myoblasts of different stages with anti-dystrophin antibody showed that purified myoblasts with the capacity of fusion can express dystrophin and can be utilized as a donating source in MTT. In the MTT experiment, eight young MDX mice were injected with normal myoblasts at a concentration of 1 x 10(6) cells. All transplated mice received daily cyclosporine A injection for immunosuppression. Two to three months later, dystrophin was found in the myoblast-transferred muscles while staining immunocytochemically. The result suggests that we successfully transferred the normal dystrophin gene from the normal myoblasts into the MDX mice since their myoblast-injected muscle could express dystrophin.


Subject(s)
Cell Transplantation , Muscles/cytology , Muscular Dystrophy, Animal/surgery , Animals , Cells, Cultured , Mice
20.
Microsurgery ; 15(12): 871-6, 1994.
Article in English | MEDLINE | ID: mdl-7707929

ABSTRACT

Following free vascularized normal muscle graft in mice, a study was made to determine whether dystrophin expression is possible in dystrophin-deficient muscles. In this study, dystrophic C57BL/10 ScSn-mdx mice were used as recipients and normal C57BL/10 ScSn mice as donors. A free vascularized quadriceps muscle 8.0 x 6.0 x 6.0 mm in size was orthotopically transplanted into a muscle defect produced in the recipient mouse. The diameter of the sutured vessels was about 0.4 mm. Transplantation was successful in 7 of 20 mice. At 12 weeks after the transplantation, the grafted muscle was examined by immunocytochemical stain using anti-dystrophin antibody. This study showed that dystrophin was expressed in the transplanted muscle but not in the adjacent recipient quadriceps muscle, suggesting that grafted donor cells with dystrophin failed to migrate into dystrophic muscle cells and fuse together. However, since the grafted normal skeletal muscle successfully survived and normal dystrophin was expressed in almost all the grafted muscle fibers, the possibility was suggested that the function of muscular dystrophy muscle can be compensated by complete replacement with a larger muscle.


Subject(s)
Muscle, Skeletal/transplantation , Muscular Dystrophy, Animal/surgery , Animals , Dystrophin/genetics , Dystrophin/metabolism , Fluorescent Antibody Technique , Gene Expression , Graft Survival , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Inbred mdx , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Sarcolemma/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...