Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Nat Commun ; 12(1): 6769, 2021 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-34819506

RESUMO

Adeno-associated virus (AAV)-mediated CRISPR-Cas9 editing holds promise to treat many diseases. The immune response to bacterial-derived Cas9 has been speculated as a hurdle for AAV-CRISPR therapy. However, immunological consequences of AAV-mediated Cas9 expression have thus far not been thoroughly investigated in large mammals. We evaluate Cas9-specific immune responses in canine models of Duchenne muscular dystrophy (DMD) following intramuscular and intravenous AAV-CRISPR therapy. Treatment results initially in robust dystrophin restoration in affected dogs but also induces muscle inflammation, and Cas9-specific humoral and cytotoxic T-lymphocyte (CTL) responses that are not prevented by the muscle-specific promoter and transient prednisolone immune suppression. In normal dogs, AAV-mediated Cas9 expression induces similar, though milder, immune responses. In contrast, other therapeutic (micro-dystrophin and SERCA2a) and reporter (alkaline phosphatase, AP) vectors result in persistent expression without inducing muscle inflammation. Our results suggest Cas9 immunity may represent a critical barrier for AAV-CRISPR therapy in large mammals.


Assuntos
Sistemas CRISPR-Cas/imunologia , Terapia Genética/efeitos adversos , Vetores Genéticos/imunologia , Músculo Esquelético/imunologia , Distrofia Muscular de Duchenne/terapia , Animais , Sistemas CRISPR-Cas/genética , Dependovirus/genética , Modelos Animais de Doenças , Cães , Distrofina/genética , Distrofina/imunologia , Edição de Genes/métodos , Genes Reporter/genética , Genes Reporter/imunologia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Humanos , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/imunologia , Distrofia Muscular de Duchenne/patologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/imunologia
2.
J Neuromuscul Dis ; 8(s2): S325-S340, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34569971

RESUMO

BACKGROUND: AAV-based gene therapy is an attractive approach to treat Duchenne muscular dystrophy (DMD) patients. Although the long-term consequences of a gene therapy approach for DMD are unknown, there is evidence in both DMD patients and animal models that dystrophin replacement by gene therapy leads to an anti-dystrophin immune response that is likely to limit the long-term use of these therapeutic strategies. OBJECTIVE: Our objective is to test whether the anti-dystrophin immune response is affected by immunomodulatory drugs in mdx mice after rAAV gene therapy. METHODS: mdx mice were treated with rAAV microdystrophin alone or in combination with immunomodulatory drugs. Dystrophin expression in skeletal muscle was assessed by mass spectrometry. Immune responses were assessed by immunophenotyping, western blot for anti-dystrophin antibodies and flow cytometry assays for antigen-specific T-cell cytokine expression. The impact on muscle was measured by grip strength assessment, in vivo torque, optical imaging for inflammation and H&E staining of sections to assess muscle damage. RESULTS: We found that AAV-9-microdystrophin gene therapy induced expression of microdystrophin, anti-dystrophin antibodies, and T-cell cytokine responses. Immunomodulatory treatments, rituximab and VBP6 completely abrogated the anti-dystrophin antibody response. Prednisolone, CTLA4-Ig, and eplerenone showed variable efficacy in blocking the anti-dystrophin immune response. In contrast, none of the drugs completely abrogated the antigen specific IFN-γ response. AAV-microdystrophin treatment significantly reduced inflammation in both forelimbs and hindlimbs, and the addition of prednisolone and VBP6 further reduced muscle inflammation. Treatment with immunomodulatory drugs, except eplerenone, enhanced the beneficial effects of AAV-microdystrophin therapy in terms of force generation. CONCLUSIONS: Our data suggest that AAV-microdystrophin treatment results in anti-dystrophin antibody and T-cell responses, and immunomodulatory treatments have variable efficacy on these responses.


Assuntos
Dependovirus/metabolismo , Distrofina/imunologia , Terapia Genética/métodos , Agentes de Imunomodulação/uso terapêutico , Distrofia Muscular de Duchenne/terapia , Animais , Expressão Gênica , Vetores Genéticos , Imunidade , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/metabolismo
3.
Proc Natl Acad Sci U S A ; 115(39): E9182-E9191, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-30181272

RESUMO

In gene therapy for Duchenne muscular dystrophy there are two potential immunological obstacles. An individual with Duchenne muscular dystrophy has a genetic mutation in dystrophin, and therefore the wild-type protein is "foreign," and thus potentially immunogenic. The adeno-associated virus serotype-6 (AAV6) vector for delivery of dystrophin is a viral-derived vector with its own inherent immunogenicity. We have developed a technology where an engineered plasmid DNA is delivered to reduce autoimmunity. We have taken this approach into humans, tolerizing to myelin proteins in multiple sclerosis and to proinsulin in type 1 diabetes. Here, we extend this technology to a model of gene therapy to reduce the immunogenicity of the AAV vector and of the wild-type protein product that is missing in the genetic disease. Following gene therapy with systemic administration of recombinant AAV6-microdystrophin to mdx/mTRG2 mice, we demonstrated the development of antibodies targeting dystrophin and AAV6 capsid in control mice. Treatment with the engineered DNA construct encoding microdystrophin markedly reduced antibody responses to dystrophin and to AAV6. Muscle force in the treated mice was also improved compared with control mice. These data highlight the potential benefits of administration of an engineered DNA plasmid encoding the delivered protein to overcome critical barriers in gene therapy to achieve optimal functional gene expression.


Assuntos
DNA , Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos , Força Muscular/genética , Distrofia Muscular de Duchenne/terapia , Plasmídeos , Animais , DNA/genética , DNA/farmacocinética , Modelos Animais de Doenças , Distrofina/genética , Distrofina/imunologia , Distrofina/metabolismo , Vetores Genéticos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos mdx , Força Muscular/imunologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/imunologia , Distrofia Muscular de Duchenne/metabolismo , Plasmídeos/genética , Plasmídeos/farmacologia
4.
Cell Transplant ; 27(7): 1096-1110, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29871519

RESUMO

Growing demonstrations of regenerative potential for some stem cells led recently to promising therapeutic proposals for neuromuscular diseases. We have shown that allogeneic MuStem cell transplantation into Golden Retriever muscular dystrophy (GRMD) dogs under continuous immunosuppression (IS) leads to persistent clinical stabilization and muscle repair. However, long-term IS in medical practice is associated with adverse effects raising safety concerns. Here, we investigate whether the IS removal or its restriction to the transplantation period could be considered. Dogs aged 4-5 months old received vascular infusions of allogeneic MuStem cells without IS (GRMDMU/no-IS) or under transient IS (GRMDMU/tr-IS). At 5 months post-infusion, persisting clinical status improvement of the GRMDMU/tr-IS dogs was observed while GRMDMU/no-IS dogs exhibited no benefit. Histologically, only 9-month-old GRMDMU/tr-IS dogs showed an increased muscle regenerative activity. A mixed cell reaction with the host peripheral blood mononucleated cells (PBMCs) and corresponding donor cells revealed undetectable to weak lymphocyte proliferation in GRMDMU/tr-IS dogs compared with a significant proliferation in GRMDMU/no-IS dogs. Importantly, any dog group showed neither cellular nor humoral anti-dystrophin responses. Our results show that transient IS is necessary and sufficient to sustain allogeneic MuStem cell transplantation benefits and prevent host immunity. These findings provide useful critical insight to designing therapeutic strategies.


Assuntos
Doenças do Cão/terapia , Terapia de Imunossupressão/métodos , Distrofia Muscular Animal/terapia , Transplante de Células-Tronco/métodos , Células Alógenas/imunologia , Animais , Cães , Distrofina/imunologia , Masculino , Distrofia Muscular Animal/imunologia , Células-Tronco/citologia , Células-Tronco/imunologia , Transplante Homólogo/métodos
5.
Hum Gene Ther ; 24(9): 797-806, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24010700

RESUMO

Duchenne muscular dystrophy (DMD) typically occurs as a result of truncating mutations in the DMD gene that result in a lack of expression of the dystrophin protein in muscle fibers. Various therapies under development are directed toward restoring dystrophin expression at the subsarcolemmal membrane, including gene transfer. In a trial of intramuscular adeno-associated virus (AAV)-mediated delivery of a therapeutic minidystrophin construct, we identified in two of six subjects the presence of a population of T cells that had been primed to recognize dystrophin epitopes before transgene delivery. As the presence of preexisting T cell immunity may have a significant effect on the success of therapeutic approaches for restoring dystrophin, we sought to determine the prevalence of such immunity within a DMD cohort from our Muscular Dystrophy Association clinic. Dystrophin-specific T cell immunity was evaluated in subjects with DMD who were either receiving the glucocorticoid steroid prednisone (n=24) or deflazacort (n=29), or who were not receiving steroids (n=17), as well as from normal age-matched control subjects (n=21). We demonstrate that increasing age correlates with an increased risk for the presence of anti-dystrophin T cell immunity, and that treatment with either corticosteroid decreases risk compared with no treatment, suggesting that steroid therapy in part may derive some of its benefit through modulation of T cell responses. The frequency of dystrophin-specific T cells detected by enzyme-linked immunospot assay was lower in subjects treated with deflazacort versus prednisone, despite similar overall corticosteroid exposure, suggesting that the effects of the two corticosteroids may not be identical in patients with DMD. T cells targeted epitopes upstream and downstream of the dystrophin gene mutation and involved the CD4⁺ helper and/or CD8⁺ cytotoxic subsets. Our data confirm the presence of preexisting circulating T cell immunity to dystrophin in a sizable proportion of patients with DMD, and emphasize the need to consider this in the design and interpretation of clinical gene therapy trials.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Distrofina/imunologia , Glucocorticoides/uso terapêutico , Distrofia Muscular de Duchenne/imunologia , Fatores Etários , Dependovirus/genética , Distrofina/genética , Terapia Genética , Humanos , Imunossupressores/uso terapêutico , Distrofia Muscular de Duchenne/tratamento farmacológico , Prednisona/uso terapêutico , Pregnenodionas/uso terapêutico
6.
Curr Gene Ther ; 12(3): 139-51, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22533379

RESUMO

The muscular dystrophies collectively represent a major health challenge, as few significant treatment options currently exist for any of these disorders. Recent years have witnessed a proliferation of novel approaches to therapy, spanning increased testing of existing and new pharmaceuticals, DNA delivery (both anti-sense oligonucleotides and plasmid DNA), gene therapies and stem cell technologies. While none of these has reached the point of being used in clinical practice, all show promise for being able to impact different types of muscular dystrophies. Our group has focused on developing direct gene replacement strategies to treat recessively inherited forms of muscular dystrophy, particularly Duchenne and Becker muscular dystrophy (DMD/BMD). Both forms of dystrophy are caused by mutations in the dystrophin gene and all cases can in theory be treated by gene replacement using synthetic forms of the dystrophin gene. The major challenges for success of this approach are the development of a suitable gene delivery shuttle, generating a suitable gene expression cassette able to be carried by such a shuttle, and achieving safe and effective delivery without elicitation of a destructive immune response. This review summarizes the current state of the art in terms of using adeno-associated viral vectors to deliver synthetic dystrophin genes for the purpose of developing gene therapy for DMD.


Assuntos
Distrofina/genética , Terapia Genética , Distrofia Muscular de Duchenne , Dependovirus , Distrofina/imunologia , Distrofina/uso terapêutico , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Imunidade Ativa/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Mutação
7.
Hum Gene Ther ; 23(2): 202-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21967249

RESUMO

Highly abbreviated micro-dystrophin genes have been intensively studied for Duchenne muscular dystrophy (DMD) gene therapy. Following adeno-associated virus (AAV) gene transfer, robust microgene expression is achieved in murine DMD models in the absence of immune suppression. Interestingly, a recent study suggests that AAV gene transfer in dystrophic dogs may require up to 18 weeks' immune suppression using a combination of three different immune-suppressive drugs (cyclosporine, mycophenolate mofetil, and anti-dog thymocyte globulin). Continued immune suppression is not only costly but also may cause untoward reactions. Further, some of the drugs (such as anti-dog thymocyte globulin) are not readily available. To overcome these limitations, we developed a novel 5-week immune suppression scheme using only cyclosporine and mycophenolate mofetil. AAV vectors (either AV.RSV.AP that expresses the heat-resistant human alkaline phosphatase gene, or AV.CMV.µDys that expresses the canine R16-17/H3/ΔC microgene) at 2.85×10(12) vg particles were injected into adult dystrophic dog limb muscles under the new immune suppression protocol. Sustained transduction was observed for nearly half year (the end of the study). The simplified immune suppression strategy described here may facilitate preclinical studies in the dog model.


Assuntos
Dependovirus/genética , Distrofina/genética , Terapia Genética , Imunossupressores/administração & dosagem , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/terapia , Animais , Soro Antilinfocitário/administração & dosagem , Ciclosporina/administração & dosagem , Modelos Animais de Doenças , Cães , Distrofina/imunologia , Feminino , Expressão Gênica , Vetores Genéticos , Humanos , Terapia de Imunossupressão , Injeções Intramusculares , Masculino , Camundongos , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/imunologia , Distrofia Muscular de Duchenne/patologia , Ácido Micofenólico/administração & dosagem , Ácido Micofenólico/análogos & derivados , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transgenes
8.
Methods Mol Biol ; 709: 39-61, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21194020

RESUMO

Several molecular approaches to Duchenne muscular dystrophy (DMD) therapy are at or near the point of clinical trial and usually involve attempts to replace the missing dystrophin protein. Although improved muscle function is the ultimate measure of success, assessment of dystrophin levels after therapy is essential to determine whether any improved function is a direct consequence of the treatment or, in the absence of improved function, to determine whether new dystrophin is present, though ineffective. The choice of a monoclonal antibody (mAb) to distinguish successful therapy from naturally occurring "revertant" fibres depends on which dystrophin exons are deleted in the DMD patient. Over the past 20 years, we have produced over 150 "exon-specific" mAbs, mapped them to different regions of dystrophin and made them available through the MDA Monoclonal Antibody Resource for research and for clinical trials tailored to individual patients. In this protocol, we describe the use of these mAb to monitor DMD gene therapy.


Assuntos
Anticorpos Monoclonais , Distrofina/análise , Distrofina/imunologia , Terapia Genética , Distrofia Muscular de Duchenne/terapia , Mapeamento de Epitopos , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Microscopia de Fluorescência , Músculos , Distrofia Muscular de Duchenne/genética
9.
N Engl J Med ; 363(15): 1429-37, 2010 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-20925545

RESUMO

We report on delivery of a functional dystrophin transgene to skeletal muscle in six patients with Duchenne's muscular dystrophy. Dystrophin-specific T cells were detected after treatment, providing evidence of transgene expression even when the functional protein was not visualized in skeletal muscle. Circulating dystrophin-specific T cells were unexpectedly detected in two patients before vector treatment. Revertant dystrophin fibers, which expressed functional, truncated dystrophin from the deleted endogenous gene after spontaneous in-frame splicing, contained epitopes targeted by the autoreactive T cells. The potential for T-cell immunity to self and nonself dystrophin epitopes should be considered in designing and monitoring experimental therapies for this disease. (Funded by the Muscular Dystrophy Association and others; ClinicalTrials.gov number, NCT00428935.).


Assuntos
Autoanticorpos/análise , Distrofina/genética , Terapia Genética , Imunidade Celular , Distrofia Muscular de Duchenne/imunologia , Linfócitos T/imunologia , Autoimunidade , Criança , DNA Viral/análise , Dependovirus , Distrofina/imunologia , Mutação da Fase de Leitura , Vetores Genéticos , Humanos , Masculino , Músculo Esquelético/química , Músculo Esquelético/imunologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Biossíntese de Proteínas , Transgenes
10.
Gene Ther ; 17(9): 1181-90, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20827278

RESUMO

Duchenne muscular dystrophy is a fatal, genetic disorder in which dystrophin-deficient muscle progressively degenerates, for which dystrophin gene transfer could provide effective treatment. The host immune response to dystrophin, however, is an obstacle to therapeutic gene expression. Understanding the dystrophin-induced host immune response will facilitate the discovery of strategies to prolong expression of recombinant dystrophin in dystrophic muscle. Using whole-body irradiation of the dystrophic mdx mouse before gene transfer, we temporally removed the immune system; a 600 rad dose removed peripheral immune cells, which were restored by self-reconstitution, and a 900 rad dose removed central and peripheral immune cells, which were restored by adoptive transfer of bone marrow from a syngeneic, dystrophin-normal donor. The anti-dystrophin humoral response was delayed and dystrophin expression was partially preserved in irradiated, vector-treated mice. Nonirradiated, vector-treated control mice lost muscle dystrophin expression completely, had an earlier anti-dystrophin humoral response and demonstrated muscle fibers focally surrounded with T cells. We conclude that dystrophin gene transfer induced anti-dystrophin humoral immunity and cell-mediated responses that were significantly diminished and delayed by temporal removal of the host central or peripheral immune cells. Furthermore, manipulation of central immunity altered the pattern of regulatory T cells in muscle.


Assuntos
Distrofina/genética , Imunidade Humoral/efeitos da radiação , Distrofia Muscular de Duchenne/imunologia , Irradiação Corporal Total , Animais , DNA Complementar/administração & dosagem , Distrofina/imunologia , Técnicas de Transferência de Genes , Vetores Genéticos , Camundongos , Camundongos Endogâmicos mdx , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/efeitos da radiação
11.
Transplantation ; 85(12): 1791-801, 2008 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-18580473

RESUMO

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.


Assuntos
Antígenos/imunologia , Transplante de Células , Distrofina/imunologia , Tolerância Imunológica/imunologia , Músculo Esquelético/citologia , Músculo Esquelético/imunologia , Animais , Transplante de Medula Óssea/imunologia , Linfócitos T CD4-Positivos/patologia , Ligante de CD40/imunologia , Antígenos CD8/imunologia , Linfócitos T CD8-Positivos/patologia , Quimera/imunologia , Modelos Animais de Doenças , Distrofina/metabolismo , Tolerância Imunológica/efeitos dos fármacos , Tolerância Imunológica/efeitos da radiação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/imunologia , Distrofia Muscular Animal/patologia , Distrofia Muscular Animal/cirurgia , Transplante de Pele/imunologia , Irradiação Corporal Total
12.
Nature ; 444(7119): 574-9, 2006 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-17108972

RESUMO

Duchenne muscular dystrophy remains an untreatable genetic disease that severely limits motility and life expectancy in affected children. The only animal model specifically reproducing the alterations in the dystrophin gene and the full spectrum of human pathology is the golden retriever dog model. Affected animals present a single mutation in intron 6, resulting in complete absence of the dystrophin protein, and early and severe muscle degeneration with nearly complete loss of motility and walking ability. Death usually occurs at about 1 year of age as a result of failure of respiratory muscles. Here we report that intra-arterial delivery of wild-type canine mesoangioblasts (vessel-associated stem cells) results in an extensive recovery of dystrophin expression, normal muscle morphology and function (confirmed by measurement of contraction force on single fibres). The outcome is a remarkable clinical amelioration and preservation of active motility. These data qualify mesoangioblasts as candidates for future stem cell therapy for Duchenne patients.


Assuntos
Células-Tronco Adultas/transplante , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Transplante de Células-Tronco , Células-Tronco Adultas/imunologia , Animais , Terapia Combinada , Creatina Quinase/sangue , Cães , Distrofina/biossíntese , Distrofina/genética , Distrofina/imunologia , Terapia Genética , Humanos , Masculino , Células Musculares , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Transplante Autólogo , Transplante Heterólogo
13.
J Neuropathol Exp Neurol ; 65(4): 371-86, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16691118

RESUMO

A clinical trial was conducted to test a new protocol of normal muscle precursor cell (MPC) allotransplantation in skeletal muscles of patients with Duchenne muscular dystrophy (DMD). Cultured MPCs obtained from one of the patient's parents were implanted in 0.25 or 1 cm of a Tibialis anterior in 9 patients with DMD. MPC injections were placed 1 to 2 mm from each other, and a similar pattern of saline injections was done in the contralateral muscle. The patients were immunosuppressed with tacrolimus. Muscle biopsies were performed at the injected sites 4 weeks later. In the biopsies of the cell-grafted sites, there were myofibers expressing donor's dystrophin in 8 patients. The percentage of myofibers expressing donor's dystrophin varied from 3.5% to 26%. Evidence of small myofiber neoformation was observed in some patients. Donor-derived dystrophin transcripts were detected by reverse transcriptase-polymerase chain reaction in the cell-grafted sites in all patients. The protocol of immunosuppression was sufficient to obtain these results, although it is not certain whether acute rejection was efficiently controlled in all the cases. In conclusion, intramuscular allotransplantation of normal MPCs can induce the expression of donor-derived dystrophin in skeletal muscles of patients with DMD, although this expression is restricted to the sites of MPC injection.


Assuntos
Distrofina/biossíntese , Células Musculares/transplante , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/terapia , Transplante de Células-Tronco , Adolescente , Animais , Criança , Distrofina/imunologia , Imunofluorescência , Rejeição de Enxerto/prevenção & controle , Antígenos de Histocompatibilidade/imunologia , Humanos , Processamento de Imagem Assistida por Computador , Imunossupressores/uso terapêutico , Hibridização in Situ Fluorescente , Camundongos , Camundongos SCID , Células Musculares/imunologia , Músculo Esquelético/imunologia , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tacrolimo/uso terapêutico
14.
Mol Ther ; 6(3): 359-68, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12231172

RESUMO

Adenoviral vector-mediated gene transfer to skeletal muscle is a promising potential treatment for Duchenne muscular dystrophy. However, the immunological response to viral antigens and the therapeutic protein expressed by the delivered gene could prevent effective treatment. In this study, we investigated the immune response induced by adenoviral and dystrophin antigens presented by high-capacity adenoviral vector-mediated dystrophin and beta-galactosidase delivery to skeletal muscle of a mouse model that is both dystrophin-deficient and lacZ transgenic. Direct intramuscular gene delivery of the high-capacity adenoviral vector encoding full-length murine dystrophin resulted in stable expression of recombinant dystrophin for 5 months in mice treated as neonates and for 4 weeks in mice treated as adults. We observed neutralizing antibody to adenoviral antigens only in mice treated as adults and not in mice treated as neonates. This suggested that adenoviral antigens were only presented at the time of vector administration when the neonatal immune system was not yet mature. In contrast, antibodies to dystrophin were observed both in mice treated as neonates and in mice treated as adults. The development of an anti-dystrophin antibody response in mice treated with the high-capacity adenoviral vector as neonates suggested that dystrophin antigens were presented to the immune system at a time remote from the gene delivery, when the immune system was mature. Interestingly, an antibody response against beta-galactosidase developed late in the course of mice treated with the high-capacity adenoviral vector as neonates, suggesting a loss of tolerance to beta-galactosidase, a self-antigen in these transgenic mice. Our results suggest that future human trials of dystrophin gene delivery will need to address the potential for immunity induced by ongoing segmental degeneration of partially treated muscle fibers and presentation of recombinant dystrophin antigens in the context of a Duchenne muscular dystrophy patient.


Assuntos
Distrofina/imunologia , Terapia Genética , Músculos/imunologia , Distrofia Muscular de Duchenne/terapia , Animais , Anticorpos Antivirais/imunologia , Dependovirus/imunologia , Distrofina/administração & dosagem , Distrofina/genética , Feminino , Vetores Genéticos/imunologia , Vetores Genéticos/farmacologia , Imunidade , Masculino , Camundongos , Camundongos Transgênicos , Músculos/efeitos dos fármacos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/imunologia
15.
J Child Neurol ; 17(5): 333-6, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12150578

RESUMO

Twelve patients from 11 Israeli families with congenital muscular dystrophy were evaluated between 1991 and 2001. There were six males and six females, of whom six were merosin negative and six were merosin positive. Serum creatine kinase levels were highly elevated in the merosin-negative group. Four of the children were cognitively normal but nonambulant. Two had unusual clinical findings of severe cognitive and motor developmental dysfunction. Four infants in the merosin-positive group who had normal serum creatine kinase levels had early-onset severe motor weakness and died within the first year of life owing to ventilatory insufficiency. The other two were ambulant and had normal cognitive development and elevated serum creatine kinase levels. Noteworthy, two of the six children with merosin-negative congenital muscular dystrophy had cognitive impairment, and four of the six children with merosin-positive congenital muscular dystrophy had a severe form of the disease with ventilatory insufficiency and death during infancy.


Assuntos
Distrofias Musculares/congênito , Distrofias Musculares/epidemiologia , Anticorpos Monoclonais/imunologia , Biópsia , Criança , Pré-Escolar , Transtornos Cognitivos/epidemiologia , Creatina Quinase/sangue , Proteínas do Citoesqueleto/deficiência , Distrofina/imunologia , Feminino , Humanos , Lactente , Israel/epidemiologia , Laminina/metabolismo , Masculino , Glicoproteínas de Membrana/deficiência , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofias Musculares/metabolismo , Insuficiência Respiratória/epidemiologia , Sarcoglicanas
16.
Eur J Cardiothorac Surg ; 21(5): 853-7, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12062274

RESUMO

BACKGROUND: While morphologic integration of transplanted fetal cardiomyocytes into the ventricular myocardium is a well-known fact, no studies have yet shown transplanted cells to coherently contribute to contraction and electrical excitation of the host myocardium. The aim of this study was to prove the hypothesis that by transplanting cardiomyocytes with a higher intrinsic rhythmic rate into the myocardium of the left ventricle, these cells could act as an ectopic pacemaker by functional coupling with host cardiomyocytes. METHODS AND RESULTS: Dissociated fetal canine atrial cardiomyocytes including sinus nodal cells were delivered into the free wall of the left ventricle of adult canine X-linked muscular dystrophy dogs (n=2). These dogs fail to express Dystrophin in both cardiac and skeletal muscle. In the control group (n=2) fetal skin fibroblasts were used for grafting. A total of 3-4 weeks after transplantation the dogs underwent catheter ablation of the atrioventricular node (AV-node) and subsequent electrophysiological mapping studies. Transplanted cells were identified by Dystrophin immunoreactivity, indicating survival and morphological integration in the recipient heart. The expression of Connexin 43 between donor and recipient cells suggested formation of gap junctions between injected and host cardiomyocytes. After catheter ablation of the AV-node, a ventricular escape rhythm emerged driving the pace of the heart and originating from the labeled transplantation site. This effect could not be observed in the control group (n=2). CONCLUSIONS: The results constitute the first observation of phenomena indicating electrical and mechanical coupling between allogeneic donor cardiomyocytes and recipient myocardium in-vivo. Further experiments are necessary to evaluate the technique as a potential therapy for atrioventricular block.


Assuntos
Transplante de Tecido Fetal , Miocárdio/citologia , Função Ventricular Esquerda/fisiologia , Animais , Nó Atrioventricular/cirurgia , Transplante de Células , Cães , Distrofina/genética , Distrofina/imunologia , Eletrocardiografia , Técnicas Eletrofisiológicas Cardíacas , Sistema de Condução Cardíaco/fisiologia , Contração Miocárdica , Função Ventricular Esquerda/genética
17.
Ann Neurol ; 49(6): 706-11, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11409421

RESUMO

Aminoglycosides have previously been shown to suppress nonsense mutations, allowing translation of full-length proteins in vitro and in animal models. In the mdx mouse, where muscular dystrophy is due to a nonsense mutation in the dystrophin gene, gentamicin suppressed truncation of the protein and ameliorated the phenotype. A subset of patients with Duchenne and Becker muscular dystrophy similarly possess a nonsense mutation, causing premature termination of dystrophin translation. Four such patients, with various stop codon sequences, were treated once daily with intravenous gentamicin at 7.5 mg/kg/day for 2 weeks. No ototoxicity or nephrotoxicity was detected. Full-length dystrophin was not detected in pre- and post-treatment muscle biopsies.


Assuntos
Códon sem Sentido/genética , Gentamicinas/uso terapêutico , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/genética , Adolescente , Biópsia , Western Blotting , Criança , Creatina Quinase/sangue , Distrofina/biossíntese , Distrofina/química , Distrofina/genética , Distrofina/imunologia , Gentamicinas/administração & dosagem , Gentamicinas/efeitos adversos , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Distrofia Muscular de Duchenne/sangue , Distrofia Muscular de Duchenne/metabolismo , Fenótipo , Resultado do Tratamento
18.
J Cell Sci ; 113 Pt 19: 3409-17, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10984432

RESUMO

The abnormal retinal neurotransmission observed in Duchenne muscular dystrophy patients has been attributed to altered expression of C-terminal products of the dystrophin gene in this tissue. Müller glial cells from rat retina express dystrophin protein Dp71, utrophin and the members of the dystrophin-associated glycoprotein complex (DGC), namely beta-dystroglycan, delta- and gamma-sarcoglycans and alpha1-syntrophin. The DGC could function in muscle as a link between the cystoskeleton and the extracellular matrix, as well as a signaling complex. However, other than in muscle the composition and intermolecular associations among members of the DGC are still unknown. Here we demonstrate that Dp71 and/or utrophin from rat retinal Müller glial cells form a complex with beta-dystroglycan, delta-sarcoglycan and alpha1-syntrophin. We also show that beta-dystroglycan is associated with alpha-dystrobrevin-1 and PSD-93 and that anti-PSD antibodies coimmunoprecipitated alpha-syntrophin with PSD-93. By overlay experiments we also found that Dp71and/or utrophin and alpha-dystroglycan from Müller cells could bind to actin and laminin, respectively. These results indicate that the DGC could have both structural and signaling functions in retina. On the basis of our accumulated evidence, we propose a hypothetical model for the molecular organization of the dystrophin-associated glycoprotein complex in retinal Müller glial cells, which would be helpful for understanding its function in the central nervous system.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas Associadas à Distrofina , Distrofina/análogos & derivados , Distrofina/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Musculares/metabolismo , Neuroglia/metabolismo , Retina/citologia , Actinas/metabolismo , Animais , Western Blotting , Química Encefálica , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Proteínas do Citoesqueleto/imunologia , Distroglicanas , Distrofina/imunologia , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular , Laminina/metabolismo , Substâncias Macromoleculares , Glicoproteínas de Membrana/imunologia , Proteínas de Membrana/imunologia , Modelos Biológicos , Proteínas Musculares/imunologia , Distrofia Muscular de Duchenne , Proteínas do Tecido Nervoso/imunologia , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/química , Testes de Precipitina , Ratos , Ratos Wistar , Retina/química , Sarcoglicanas , Utrofina
19.
J Cell Sci ; 113 ( Pt 15): 2715-24, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10893187

RESUMO

Dystrophin is the key component in the assembly and maintenance of the dystrophin-associated protein complex (DPC) in skeletal muscle. In kidney, dystroglycan, an integral component of the DPC, is involved in kidney epithelial morphogenesis, suggesting that the DPC is important in linking the extracellular matrix to the internal cytoskeleton of kidney epithelia. Here, we have investigated the molecular architecture of dystrophin-like protein complexes in kidneys from normal and dystrophin-deficient mice. Using isoform-specific antibodies, we show that the different cell types that make up the kidney maintain different dystrophin-like complexes. These complexes can be broadly grouped according to their dystrobrevin content: beta-dystrobrevin containing complexes are present at the basal region of renal epithelial cells, whilst alpha-dystrobrevin-1 containing complexes are found in endothelial and smooth muscle cells. Furthermore, these complexes are maintained even in the absence of all dystrophin isoforms. Thus our data suggest that the functions and assembly of the dystrophin-like complexes in kidney differ from those in skeletal muscle and implicate a protein other than dystrophin as the primary molecule in the assembly and maintenance of kidney complexes. Our findings also provide a possible explanation for the lack of kidney pathology in Duchenne muscular dystrophy patients and mice lacking all dystrophin isoforms.


Assuntos
Proteínas Associadas à Distrofina , Rim/química , Rim/metabolismo , Proteínas de Membrana/análise , Proteínas de Membrana/metabolismo , Animais , Especificidade de Anticorpos , Proteínas do Citoesqueleto/análise , Proteínas do Citoesqueleto/imunologia , Proteínas do Citoesqueleto/metabolismo , Distrofina/análogos & derivados , Distrofina/análise , Distrofina/imunologia , Distrofina/metabolismo , Células Epiteliais/química , Células Epiteliais/metabolismo , Imunofluorescência , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Proteínas Musculares/análise , Proteínas Musculares/imunologia , Proteínas Musculares/metabolismo , Néfrons/química , Néfrons/metabolismo , Utrofina
20.
Muscle Nerve ; 21(11): 1549-53, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9771686

RESUMO

An 8-year-old girl developed weakness over 2 years and an elevated creatine kinase. The biopsy was most consistent with an active dystrophy with many inflammatory cells present. A trial of immunosuppression was started. In the first 2 months of treatment with prednisone, she had functionally and quantitatively significant improvement in her proximal strength. Over 3 years of treatment she maintained stable strength. Subsequent genetic studies showed that she had primary alpha-sarcoglycan deficiency. The timing and the degree of benefit in strength were similar to that seen in boys with Duchenne muscular dystrophy who are treated with prednisone.


Assuntos
Anti-Inflamatórios/administração & dosagem , Proteínas do Citoesqueleto/deficiência , Glicoproteínas de Membrana/deficiência , Distrofias Musculares/tratamento farmacológico , Distrofias Musculares/metabolismo , Prednisona/administração & dosagem , Substituição de Aminoácidos , Anticorpos , Biópsia , Criança , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/imunologia , Análise Mutacional de DNA , Distrofina/análise , Distrofina/imunologia , Feminino , Humanos , Imunoglobulinas/análise , Terapia de Imunossupressão , Macrófagos/imunologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Músculo Esquelético/química , Músculo Esquelético/imunologia , Músculo Esquelético/patologia , Distrofias Musculares/genética , Mutação Puntual , Sarcoglicanas , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA