Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
1.
Int J Mol Sci ; 25(11)2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38892308

RESUMO

Sarcospan (SSPN) is a 25-kDa transmembrane protein that is broadly expressed at the cell surface of many tissues, including, but not limited to, the myofibers from skeletal and smooth muscles, cardiomyocytes, adipocytes, kidney epithelial cells, and neurons. SSPN is a core component of the dystrophin-glycoprotein complex (DGC) that links the intracellular actin cytoskeleton with the extracellular matrix. It is also associated with integrin α7ß1, the predominant integrin expressed in skeletal muscle. As a tetraspanin-like protein with four transmembrane spanning domains, SSPN functions as a scaffold to facilitate protein-protein interactions at the cell membrane. Duchenne muscular dystrophy, Becker muscular dystrophy, and X-linked dilated cardiomyopathy are caused by the loss of dystrophin at the muscle cell surface and a concomitant loss of the entire DGC, including SSPN. SSPN overexpression ameliorates Duchenne muscular dystrophy in the mdx murine model, which supports SSPN being a viable therapeutic target. Other rescue studies support SSPN as a biomarker for the proper assembly and membrane expression of the DGC. Highly specific and robust antibodies to SSPN are needed for basic research on the molecular mechanisms of SSPN rescue, pre-clinical studies, and biomarker evaluations in human samples. The development of SSPN antibodies is challenged by the presence of its four transmembrane domains and limited antigenic epitopes. To address the significant barrier presented by limited commercially available antibodies, we aimed to generate a panel of robust SSPN-specific antibodies that can serve as a resource for the research community. We created antibodies to three SSPN protein epitopes, including the intracellular N- and C-termini as well as the large extracellular loop (LEL) between transmembrane domains 3 and 4. We developed a panel of rabbit antibodies (poly- and monoclonal) against an N-terminal peptide fragment of SSPN. We used several assays to show that the rabbit antibodies recognize mouse SSPN with a high functional affinity and specificity. We developed mouse monoclonal antibodies against the C-terminal peptide and the large extracellular loop of human SSPN. These antibodies are superior to commercially available antibodies and outperform them in various applications, including immunoblotting, indirect immunofluorescence analysis, immunoprecipitation, and an ELISA. These newly developed antibodies will significantly improve the quality and ease of SSPN detection for basic and translational research.


Assuntos
Proteínas de Membrana , Pesquisa Translacional Biomédica , Animais , Humanos , Camundongos , Distrofina/metabolismo , Distrofina/imunologia , Distrofina/genética , Integrinas/metabolismo , Integrinas/imunologia , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Distrofia Muscular de Duchenne/imunologia , Distrofia Muscular de Duchenne/metabolismo
3.
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
4.
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
5.
Sci Rep ; 11(1): 1128, 2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33441839

RESUMO

Emerging and promising therapeutic interventions for Duchenne muscular dystrophy (DMD) are confounded by the challenges of quantifying dystrophin. Current approaches have poor precision, require large amounts of tissue, and are difficult to standardize. This paper presents an immuno-mass spectrometry imaging method using gadolinium (Gd)-labeled anti-dystrophin antibodies and laser ablation-inductively coupled plasma-mass spectrometry to simultaneously quantify and localize dystrophin in muscle sections. Gd is quantified as a proxy for the relative expression of dystrophin and was validated in murine and human skeletal muscle sections following k-means clustering segmentation, before application to DMD patients with different gene mutations where dystrophin expression was measured up to 100 µg kg-1 Gd. These results demonstrate that immuno-mass spectrometry imaging is a viable approach for pre-clinical to clinical research in DMD. It rapidly quantified relative dystrophin in single tissue sections, efficiently used valuable patient resources, and may provide information on drug efficacy for clinical translation.


Assuntos
Distrofina/análise , Distrofia Muscular de Duchenne/metabolismo , Músculo Quadríceps/química , Adolescente , Idoso de 80 Anos ou mais , Animais , Criança , Distrofina/genética , Distrofina/imunologia , Feminino , Imunofluorescência , Gadolínio , Humanos , Imuno-Histoquímica , Masculino , Espectrometria de Massas , Camundongos , Fibras Musculares Esqueléticas/química , Distrofia Muscular de Duchenne/genética , Mutação
6.
J Immunol ; 203(2): 476-484, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31142604

RESUMO

Eosinophils are present in muscle lesions associated with Duchenne muscular dystrophy and dystrophin-deficient mdx mice that phenocopy this disorder. Although it has been hypothesized that eosinophils promote characteristic inflammatory muscle damage, this has not been fully examined. In this study, we generated mice with the dystrophin mutation introduced into PHIL, a strain with a transgene that directs lineage-specific eosinophil ablation. We also explored the impact of eosinophil overabundance on dystrophinopathy by introducing the dystrophin mutation into IL-5 transgenic mice. We evaluated the degree of eosinophil infiltration in association with myofiber size distribution, centralized nuclei, serum creatine kinase, and quantitative histopathology scores. Among our findings, eosinophils were prominent in the quadriceps muscles of 4-wk-old male mdx mice but no profound differences were observed in the quantitative measures of muscle damage when comparing mdx versus mdx.PHIL versus mdx.IL5tg mice, despite dramatic differences in eosinophil infiltration (CD45+CD11c-Gr1-MHC class IIloSiglecF+ eosinophils at 1.2 ± 0.34% versus <0.1% versus 20 ± 7.6% of total cells, respectively). Further evaluation revealed elevated levels of eosinophil chemoatttractants eotaxin-1 and RANTES in the muscle tissue of all three dystrophin-deficient strains; eotaxin-1 concentration in muscle correlated inversely with age. Cytokines IL-4 and IL-1R antagonist were also detected in association with eosinophils in muscle. Taken together, our findings challenge the long-held perception of eosinophils as cytotoxic in dystrophin-deficient muscle; we show clearly that eosinophil infiltration is not a driving force behind acute muscle damage in the mdx mouse strain. Ongoing studies will focus on the functional properties of eosinophils in this unique microenvironment.


Assuntos
Eosinófilos/imunologia , Distrofia Muscular de Duchenne/imunologia , Animais , Modelos Animais de Doenças , Distrofina/imunologia , Feminino , Interleucina-4/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Músculo Esquelético/imunologia , Receptores de Interleucina-1/imunologia
7.
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
8.
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
9.
PLoS One ; 9(2): e88280, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24516626

RESUMO

Epitope-specific monoclonal antibodies can provide unique insights for studying cellular proteins. Dystrophin is one of the largest cytoskeleton proteins encoded by 79 exons. The absence of dystrophin results in Duchenne muscular dystrophy (DMD). Over the last two decades, dozens of exon-specific human dystrophin monoclonal antibodies have been developed and successfully used for DMD diagnosis. Unfortunately, the majority of these antibodies have not been thoroughly characterized in dystrophin-deficient dogs, an outstanding large animal model for translational research. To fill the gap, we performed a comprehensive study on 65 dystrophin monoclonal antibodies in normal and dystrophic dogs (heart and skeletal muscle) by immunofluorescence staining and western blot. For comparison, we also included striated muscles from normal BL10 and dystrophin-null mdx mice. Our analysis revealed distinctive species, tissue and assay-dependent recognition patterns of different antibodies. Importantly, we identified 15 antibodies that can consistently detect full-length canine dystrophin in both immunostaining and western blot. Our results will serve as an important reference for studying DMD in the canine model.


Assuntos
Anticorpos Monoclonais/metabolismo , Distrofina/imunologia , Epitopos/imunologia , Distrofia Muscular Animal/imunologia , Distrofia Muscular de Duchenne/imunologia , Animais , Western Blotting , Modelos Animais de Doenças , Cães , Éxons , Imuno-Histoquímica , Camundongos , Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Duchenne/metabolismo
10.
Neuromuscul Disord ; 24(3): 195-200, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24361045

RESUMO

Most pathogenic mutations in Duchenne and Becker muscular dystrophies involve deletion of single or multiple exons from the dystrophin gene, so exon-specific monoclonal antibodies (mAbs) can be used to distinguish normal and mutant dystrophin proteins. In Duchenne therapy trials, mAbs can be used to identify or rule out dystrophin-positive "revertant" fibres, which have an internally-deleted dystrophin protein and which occur naturally in some Duchenne patients. Using phage-displayed peptide libraries, we now describe the new mapping of the binding sites of five dystrophin mAbs to a few amino-acids within single exons. The phage display method also confirmed previous mapping of MANEX1A (exon 1) and MANDRA1 (exon 77) by other methods. Of the 79 dystrophin exons, mAbs are now available against single exons 1, 6, 8, 12, 13, 14, 17, 21, 26, 28, 38, 41, 43, 44, 45, 46, 47, 50, 51, 58, 59, 62, 63, 75 and 77. Many have been used in clinical trials, as well as for diagnosis and studies of dystrophin isoforms.


Assuntos
Anticorpos Monoclonais/imunologia , Distrofina/imunologia , Distrofina/química , Mapeamento de Epitopos , Humanos , Masculino , Distrofia Muscular de Duchenne/imunologia , Distrofia Muscular de Duchenne/terapia , Análise de Sequência de Proteína
11.
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
12.
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
13.
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
14.
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
15.
Patol Fiziol Eksp Ter ; (4): 33-7, 2011.
Artigo em Russo | MEDLINE | ID: mdl-22359931

RESUMO

Dystrophin is a protein linking the cytoskeleton to a complex of transmembrane proteins that interact with the extracellular matrix. The fragility of the cardiomyocyte cell membrane resulting from the lack of dystrophin is thought to cause an excessive susceptibility to mechanical stress. Based on surface ECC we demonstrate the differences of cardiac phenotype in young (2- to 3-mo-old) and aged (over 1,5 years) dystrophin-deficient mdx mouse and normal mouse with the same genetic background. It was shown that main alterations in the mdx electrocardiogram concern primarily ventricular conduction velocity (QRS complex duration) and time of ventricular repolarization (QT interval) duration).The issue under discussion is whether dystrophin deficient mdx model can be used in research studies in cardioimmunology.


Assuntos
Distrofina/imunologia , Miocárdio/imunologia , Estresse Fisiológico/imunologia , Envelhecimento/genética , Envelhecimento/imunologia , Animais , Citoesqueleto/genética , Citoesqueleto/imunologia , Citoesqueleto/metabolismo , Distrofina/genética , Distrofina/metabolismo , Eletrocardiografia , Feminino , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos mdx , Miocárdio/metabolismo , Fenótipo , Estresse Fisiológico/genética
16.
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
18.
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
19.
Int Rev Immunol ; 27(5): 375-87, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18853344

RESUMO

Signaling through nuclear factor-kappa B (NF-kappaB) is emerging as an important regulator of muscle development, maintenance, and regeneration. Classic signaling modulates early muscle development by enhancing proliferation and inhibiting differentiation, and alternative signaling promotes myofiber maintenance and metabolism. Likewise, NF-kappaB signaling is critical for the development of immunity. Although these processes occur normally, dysregulation of NF-kappaB signaling has prohibitive effects on muscle growth and regeneration and can perpetuate inflammation in muscle diseases. Aberrant NF-kappaB signaling from immune and muscle cells has been detected and implicated in the pathologic progression of numerous dystrophies and myopathies, indicating that targeted NF-kappaB inhibitors may prove clinically beneficial.


Assuntos
Distrofina/metabolismo , Doenças Musculares/imunologia , Distrofias Musculares/imunologia , NF-kappa B/metabolismo , Animais , Curcumina/farmacologia , Distrofina/genética , Distrofina/imunologia , Glucocorticoides/farmacologia , Humanos , Quinase I-kappa B/metabolismo , Inflamação , Camundongos , Camundongos Endogâmicos mdx , Desenvolvimento Muscular/imunologia , Doenças Musculares/metabolismo , Doenças Musculares/terapia , Distrofias Musculares/metabolismo , Distrofias Musculares/terapia , NF-kappa B/imunologia , Ratos , Transdução de Sinais , Ativação Transcricional
20.
J Histochem Cytochem ; 56(11): 969-75, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18645208

RESUMO

Antigen detection with indirect immunohistochemical methods is hampered by high background staining if the primary antibody is from the same species as the examined tissue. This high background can be eliminated in unfixed cryostat sections of mouse skeletal muscle by boiling sections in PBS, and several proteins including even the low abundant dystrophin protein can then be easily detected with murine monoclonal antibodies. However, not all antigens withstand the boiling procedure. Immunoreactivity of some of these antigens can be restored by subsequent washing in Triton X-100, whereas immunoreactivity of other proteins is not restored by this detergent treatment. When such thermolabile proteins are labeled with polyclonal primary antibodies followed by dichlorotriazinylaminofluorescein-conjugated secondary antibodies and boiled, the fluorescence signal persists, and sections can then be processed with a monoclonal antibody for double immunostaining of a protein unaffected by boiling. This stability of certain fluorochromes on heating can also be exploited for double immunofluorescence labeling of two different thermostable proteins with murine monoclonal antibodies as well as for combination with Y-chromosome fluorescence in situ hybridization. Our method should extend the range of monoclonal antibodies applicable to tissues derived from the same species as the monoclonal antibodies.


Assuntos
Anticorpos Monoclonais , Antígenos/metabolismo , Músculo Esquelético/metabolismo , Animais , Antígenos/imunologia , Soluções Tampão , Distrofina/imunologia , Distrofina/metabolismo , Secções Congeladas , Temperatura Alta , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Masculino , Camundongos , Camundongos Endogâmicos mdx , Fosfatos , Cloreto de Sódio , Soluções , Cromossomo Y
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...