Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Neurology ; 78(12): 904-13, 2012 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-22402864

RESUMEN

OBJECTIVE: To determine whether pentoxifylline (PTX) slows the decline of muscle strength and function in ambulatory boys with Duchenne muscular dystrophy (DMD). METHODS: This was a multicenter, randomized, double-blinded, controlled trial comparing 12 months of daily treatment with PTX or placebo in corticosteroid-treated boys with DMD using a slow-release PTX formulation (~20 mg/kg/day). The primary outcome was the change in mean total quantitative muscle testing (QMT) score. Secondary outcomes included changes in QMT subscales, manual muscle strength, pulmonary function, and timed function tests. Outcomes were compared using Student t tests and a linear mixed-effects model. Adverse events (AEs) were compared using the Fisher exact test. RESULTS: A total of 64 boys with DMD with a mean age of 9.9 ± 2.9 years were randomly assigned to PTX or placebo in 11 participating Cooperative International Neuromuscular Research Group centers. There was no significant difference between PTX and the placebo group in total QMT scores (p = 0.14) or in most of the secondary outcomes after a 12-month treatment. The use of PTX was associated with mild to moderate gastrointestinal or hematologic AEs. CONCLUSION: The addition of PTX to corticosteroid-treated boys with DMD at a moderate to late ambulatory stage of disease did not improve or halt the deterioration of muscle strength and function over a 12-month study period. CLASSIFICATION OF EVIDENCE: This study provides Class I evidence that treatment with PTX does not prevent deterioration in muscle function or strength in corticosteroid-treated boys with DMD.


Asunto(s)
Distrofia Muscular de Duchenne/tratamiento farmacológico , Pentoxifilina/uso terapéutico , Inhibidores de Fosfodiesterasa/uso terapéutico , Corticoesteroides/uso terapéutico , Niño , Preparaciones de Acción Retardada , Progresión de la Enfermedad , Método Doble Ciego , Quimioterapia Combinada , Humanos , Masculino , Fuerza Muscular/fisiología , Distrofia Muscular de Duchenne/fisiopatología , Distrofia Muscular de Duchenne/psicología , Examen Neurológico , Pentoxifilina/administración & dosificación , Pentoxifilina/efectos adversos , Inhibidores de Fosfodiesterasa/administración & dosificación , Inhibidores de Fosfodiesterasa/efectos adversos , Calidad de Vida , Pruebas de Función Respiratoria , Tamaño de la Muestra , Resultado del Tratamiento
2.
Neurology ; 77(5): 444-52, 2011 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-21753160

RESUMEN

OBJECTIVE: To perform a double-blind, randomized study comparing efficacy and safety of daily and weekend prednisone in boys with Duchenne muscular dystrophy (DMD). METHODS: A total of 64 boys with DMD who were between 4 and 10 years of age were randomized at 1 of 12 centers of the Cooperative International Neuromuscular Research Group. Efficacy and safety of 2 prednisone schedules (daily 0.75 mg/kg/day and weekend 10 mg/kg/wk) were evaluated over 12 months. RESULTS: Equivalence was met for weekend and daily dosing of prednisone for the primary outcomes of quantitative muscle testing (QMT) arm score and QMT leg score. Secondary strength scores for QMT elbow flexors also showed equivalence between the 2 treatment groups. Overall side effect profiles of height and weight, bone density, cataract formation, blood pressure, and behavior, analyzed at 12 months, did not differ between weekend and daily dosing of prednisone. CONCLUSIONS: Weekend dosing of prednisone is equally beneficial to the standard daily dosing of prednisone. Analysis of side effect profiles demonstrated overall tolerability of both dosing regimens. CLASSIFICATION OF EVIDENCE: This study provides Class I evidence that weekend prednisone dosing is as safe and effective as daily prednisone in preserving muscle strength and preventing body mass index increases in boys with DMD over a 12-month period.


Asunto(s)
Glucocorticoides/administración & dosificación , Distrofia Muscular de Duchenne/tratamiento farmacológico , Prednisona/administración & dosificación , Factores de Edad , Índice de Masa Corporal , Niño , Preescolar , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Esquema de Medicación , Estudios de Seguimiento , Humanos , Masculino , Fuerza Muscular/efectos de los fármacos , Distrofia Muscular de Duchenne/fisiopatología , Resultado del Tratamiento
3.
Gene Ther ; 17(9): 1181-90, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20827278

RESUMEN

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.


Asunto(s)
Distrofina/genética , Inmunidad Humoral/efectos de la radiación , Distrofia Muscular de Duchenne/inmunología , Irradiación Corporal Total , Animales , ADN Complementario/administración & dosificación , Distrofina/inmunología , Técnicas de Transferencia de Gen , Vectores Genéticos , Ratones , Ratones Endogámicos mdx , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/efectos de la radiación
4.
Gene Ther ; 17(12): 1476-83, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20720575

RESUMEN

The IκB kinase (IKKα, ß and the regulatory subunit IKKγ) complex regulates nuclear factor of κB (NF-κB) transcriptional activity, which is upregulated in many chronic inflammatory diseases. NF-κB signaling promotes inflammation and limits muscle regeneration in Duchenne muscular dystrophy (DMD), resulting in fibrotic and fatty tissue replacement of muscle that exacerbates the wasting process in dystrophic muscles. Here, we examined whether dominant-negative forms of IKKα (IKKα-dn) and IKKß (IKKß-dn) delivered by adeno-associated viral (AAV) vectors to the gastrocnemius (GAS) and tibialis anterior (TA) muscles of 1, 2 and 11-month-old mdx mice, a murine DMD model, block NF-κB activation and increase muscle regeneration. At 1 month post-treatment, the levels of nuclear NF-κB in locally treated muscle were decreased by gene transfer with either AAV-CMV-IKKα-dn or AAV-CMV-IKKß-dn, but not by IKK wild-type controls (IKKα and ß) or phosphate-buffered saline (PBS). Although treatment with AAV-IKKα-dn or AAV-IKKß-dn vectors had no significant effect on muscle regeneration in young mdx mice treated at 1 and 2 months of age and collected 1 month later, treatment of old (11 months) mdx with AAV-CMV-IKKα-dn or AAV-CMV-IKKß-dn significantly increased levels of muscle regeneration. In addition, there was a significant decrease in myofiber necrosis in the AAV-IKKα-dn- and AAV-IKKß-dn-treated mdx muscle in both young and old mice. These results demonstrate that inhibition of IKKα or IKKß in dystrophic muscle reduces the adverse effects of NF-κB signaling, resulting in a therapeutic effect. Moreover, these results clearly demonstrate the therapeutic benefits of inhibiting NF-κB activation by AAV gene transfer in dystrophic muscle to promote regeneration, particularly in older mdx mice, and block necrosis.


Asunto(s)
Dependovirus/genética , Terapia Genética , Quinasa I-kappa B , Músculo Esquelético/fisiología , Distrofia Muscular de Duchenne , FN-kappa B , Animales , Núcleo Celular/enzimología , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/enzimología , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/enzimología , Distrofia Muscular de Duchenne/terapia , FN-kappa B/genética , FN-kappa B/metabolismo , Regeneración/fisiología , Transducción de Señal/genética
5.
Gene Ther ; 17(11): 1355-62, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20535217

RESUMEN

Duchenne muscular dystrophy (DMD) is a devastating primary muscle disease with pathological changes in skeletal muscle that are ongoing at the time of birth. Progressive deterioration in striated muscle function in affected individuals ultimately results in early death due to cardio-pulmonary failure. As affected individuals can be identified before birth by prenatal genetic testing for DMD, gene replacement treatment can be started in utero. This approach offers the possibility of preventing pathological changes in muscle that begin early in life. To test in utero gene transfer in the mdx mouse model of DMD, a minidystrophin gene driven by the human cytomegalovirus promoter was delivered systemically by an intraperitoneal injection to the fetus at embryonic day 16. Treated mdx mice studied at 9 weeks after birth showed widespread expression of recombinant dystrophin in skeletal muscle, restoration of the dystrophin-associated glycoprotein complex in dystrophin-expressing muscle fibers, improved muscle pathology, and functional benefit to the transduced diaphragm compared with untreated littermate controls. These results support the potential of the AAV8 vector to efficiently cross the blood vessel barrier to achieve systemic gene transfer to skeletal muscle in utero in a mouse model of muscular dystrophy, to significantly improve the dystrophic phenotype and to ameliorate the processes that lead to exhaustion of the skeletal muscle regenerative capacity.


Asunto(s)
Distrofina/genética , Terapia Genética , Distrofia Muscular de Duchenne/terapia , Animales , Citomegalovirus/genética , Dependovirus/genética , Distrofina/metabolismo , Vectores Genéticos/administración & dosificación , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/genética , Fenotipo , Regiones Promotoras Genéticas
6.
Gene Ther ; 16(9): 1130-7, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19474806

RESUMEN

One of the major challenges in the treatment of primary muscle disorders, which often affect many muscle groups, is achieving efficient, widespread transgene expression in muscle. In utero gene transfer can potentially address this problem by accomplishing the gene delivery when the tissue mass is small and the immune system is immature. Earlier studies with systemic in utero adeno-associated viral (AAV) vector serotype 1 gene delivery to embryonic day 16 (E-16) pups resulted in high levels of transduction in diaphragm and intercostal muscles, but no detectable transgene expression in limb muscles. Recently, newer AAV serotypes, such as AAV8, have shown widespread and high transgene expression in skeletal muscles and diaphragm by systemic delivery in adult and neonatal mice. We tested AAV8 vector gene delivery by intraperitoneal administration in E-16 mice in utero. Using an AAV8 vector carrying a lacZ reporter gene, we observed high-level transduction of diaphragm and intercostal muscles and more moderate transduction of multiple limb muscles and heart. Our current studies show the potential of AAV8 to achieve widespread muscle transduction in utero and suggest its therapeutic potential for primary muscle disorders.


Asunto(s)
Dependovirus/genética , Diafragma/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Músculo Esquelético/metabolismo , Animales , Dependovirus/clasificación , Diafragma/embriología , Femenino , Expresión Génica , Genes Reporteros , Terapia Genética/métodos , Vectores Genéticos/farmacocinética , Inyecciones Intraperitoneales , Operón Lac , Ratones , Fibras Musculares de Contracción Rápida/metabolismo , Músculo Esquelético/embriología , Enfermedades Musculares/terapia , Embarazo , Distribución Tisular , Transducción Genética , beta-Galactosidasa/metabolismo
7.
Gene Ther ; 15(7): 531-6, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18273052

RESUMEN

In utero gene therapy for genetic diseases, such as muscular dystrophies, offers potential advantages over postnatal treatment including vector delivery at the earliest point in the disease and treatment prior to full maturation of the immune system. This study examines in utero gene delivery of full-length murine dystrophin to the murine mdx model for Duchenne muscular dystrophy using a high-capacity adenoviral vector. We examined dystrophin expression, spread of vector, morphology and specific force production of the tibialis anterior muscle 9 weeks after intramuscular in utero injection. Recombinant dystrophin was expressed in the hindlimb muscles, with the majority of animals having expression in two muscles of the injected hindlimb. The dystrophin-glycoprotein complex was restored in those muscle fibers expressing recombinant dystrophin. Analysis of the percentage of dystrophin-expressing muscle fibers with centrally placed nuclei revealed effective protection from cycles of degeneration and regeneration normally seen in muscle fibers lacking dystrophin. However, due to low levels of muscle gene transfer, further advances in the efficiency of adenoviral vector-mediated gene delivery would be required for clinical applications of in utero gene therapy for primary myopathies such as Duchenne muscular dystrophy.


Asunto(s)
Distrofina/genética , Terapias Fetales/métodos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Distrofia Muscular de Duchenne/terapia , Adenoviridae/genética , Animales , Animales Recién Nacidos , Distrofina/análisis , Distrofina/metabolismo , Femenino , Expresión Génica , Vectores Genéticos/genética , Miembro Posterior , Inyecciones Intramusculares , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Animal , Distrofia Muscular de Duchenne/embriología , Distrofia Muscular de Duchenne/patología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción Genética/métodos
8.
Gene Ther ; 12(1): 39-47, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15483668

RESUMEN

In utero gene delivery could offer the advantage of treatment at an early stage for genetic disorders such as Duchenne muscular dystrophy (DMD) in which the inevitable process of muscle degeneration is already initiated at birth. Furthermore, treatment of fetal muscle with adenoviral (Ad) vectors is attractive because of a high density of Ad receptors, easy vector accessibility due to immaturity of the basal lamina and the possibility of treating stem cells. Previously, we demonstrated the efficient transduction of fetal muscle by high-capacity Ad (HC-Ad) vectors. In this study, we compared HC-Ad and first-generation Ad (FG-Ad) vectors for longevity of lacZ transgene expression, toxicity and induction of immunity after direct vector-mediated in utero gene delivery to fetal C57BL/6 mice muscle 16 days after conception (E-16). The total amount of beta-galactosidase (betagal) expressed from the HC-Ad vector remained stable for the 5 months of the study, although the concentration of betagal decreased due to muscle growth. Higher survival rates that reflect lower levels of toxicity were observed in those mice transduced with an HC-Ad vector as compared to an FG-Ad vector. The toxicity induced by FG-Ad vector gene delivery was dependent on mouse strain and vector dose. Animals treated with either HC-Ad and FG-Ad vectors developed non-neutralizing antibodies against Ad capsid and antibodies against betagal, but these antibodies did not cause loss of vector genomes from transduced muscle. In a mouse model of DMD, dystrophin gene transfer to muscle in utero using an HC-Ad vector restored the dystrophin-associated glycoproteins. Our results demonstrate that long-term transgene expression can be achieved by HC-Ad vector-mediated gene delivery to fetal muscle, although strategies of vector integration may need to be considered to accommodate muscle growth.


Asunto(s)
Adenoviridae/genética , Enfermedades Fetales/terapia , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Músculo Esquelético/embriología , Distrofia Muscular de Duchenne/terapia , Animales , Distrofina/genética , Expresión Génica , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Transgenes
9.
Gene Ther ; 11(19): 1453-61, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15269713

RESUMEN

Adenoviral (Ad) vector-mediated gene delivery of normal, full-length dystrophin to skeletal muscle provides a promising strategy for the treatment of Duchenne muscular dystrophy (DMD), an X-linked recessive, dystrophin-deficient muscle disease. Studies in animal models suggest that successful DMD gene therapy by Ad vector-mediated gene transfer would be precluded by cellular and humoral immune responses induced by vector capsid and transgene proteins. To address the immunity induced by Ad vector-mediated dystrophin gene delivery to dystrophic muscle, we developed high-capacity adenoviral (HC-Ad) vectors expressing mouse dystrophin driven by the muscle creatine kinase promoter (AdmDys) and mCTLA4Ig (AdmCTLA4Ig) individually, or together from one vector (AdmCTLA4Ig/mDys). We found stable expression of dystrophin protein in the tibialis anterior muscles of mdx mice, coinjected with AdmCTLA4Ig and AdmDys, or injected alone with AdmCTLA4Ig/mDys, whereas the expression of dystrophin protein in the control group coinjected with AdmDys and an empty vector decreased by at least 50% between 2 and 8 weeks after administration. Additionally, we observed reductions in Ad vector-induced Th1 and Th2 cytokines, Ad vector-specific cytotoxic T lymphocyte activation and neutralizing anti-Ad antibodies in both experimental groups that received a mCTLA4Ig-expressing vector as compared to the control group. This study demonstrates that the coexpression of mCTLA4Ig and dystrophin in skeletal muscle provided by HC-Ad vector-mediated gene transfer can provide stable expression of dystrophin in immunocompetent, adult mdx mouse muscle and applies a potentially powerful strategy to overcome adaptive immunity induced by Ad vector-mediated dystrophin gene delivery toward the ultimate goal of treatment for DMD.


Asunto(s)
Adenoviridae/genética , Distrofina/genética , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Inmunoconjugados/genética , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/terapia , Transducción Genética/métodos , Abatacept , Adenoviridae/inmunología , Animales , Anticuerpos Antivirales/inmunología , Citocinas/inmunología , Distrofina/metabolismo , Expresión Génica , Ingeniería Genética , Vectores Genéticos/inmunología , Inmunoconjugados/sangre , Activación de Linfocitos , Ratones , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/sangre , Distrofia Muscular de Duchenne/inmunología , Linfocitos T Citotóxicos/inmunología
10.
Gene Ther ; 10(21): 1821-9, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12960972

RESUMEN

High levels of alpha(v) integrin expression by fetal muscle suggested that vector re-targeting to integrins could enhance adenoviral vector-mediated transduction, thereby increasing safety and efficacy of muscle gene transfer in utero. High-capacity adenoviral (HC-Ad) vectors modified by an Arg-Gly-Asp (RGD) peptide motif in the HI loop of the adenoviral fiber (RGD-HC-Ad) have demonstrated efficient gene transfer through binding to alpha(v) integrins. To test integrin targeting of HC-Ad vectors for fetal muscle gene transfer, we compared unmodified and RGD-modified HC-Ad vectors. In vivo, unmodified HC-Ad vector transduced fetal mouse muscle with four-fold higher efficiency compared to RGD-HC-Ad vector. Confirming that the difference was due to muscle cell autonomous factors and not mechanical barriers, transduction of primary myogenic cells isolated from murine fetal muscle in vitro demonstrated a three-fold better transduction by HC-Ad vector than by RGD-HC-Ad vector. We hypothesized that the high expression level of coxsackievirus and adenovirus receptor (CAR), demonstrated in fetal muscle cells both in vitro and in vivo, was the crucial variable influencing the relative transduction efficiencies of HC-Ad and RGD-HC-Ad vectors. To explore this further, we studied transduction by HC-Ad and RGD-HC-Ad vectors in paired cell lines that expressed alpha(v) integrins and differed only by the presence or absence of CAR expression. The results increase our understanding of factors that will be important for retargeting HC-Ad vectors to enhance gene transfer to fetal muscle.


Asunto(s)
Adenoviridae/genética , Enfermedades Fetales/terapia , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Músculo Esquelético/embriología , Oligopéptidos/genética , Secuencias de Aminoácidos , Animales , Femenino , Expresión Génica , Marcación de Gen , Integrinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Embarazo , Transducción Genética/métodos , beta-Galactosidasa/genética
11.
Proc Natl Acad Sci U S A ; 98(24): 14126-31, 2001 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-11717466

RESUMEN

Mitochondrial nitric oxide synthase (mtNOS), its cellular NOS isoform, and the effects of mitochondrially produced NO on bioenergetics have been controversial since mtNOS was first proposed in 1995. Here we functionally demonstrate the presence of a NOS in cardiac mitochondria. This was accomplished by direct porphyrinic microsensor measurement of Ca(2+)-dependent NO production in individual mitochondria isolated from wild-type mouse hearts. This NO production could be inhibited by NOS antagonists or protonophore collapse of the mitochondrial membrane potential. The similarity of mtNOS to the neuronal isoform was deduced by the absence of NO production in the mitochondria of knockout mice for the neuronal, but not the endothelial or inducible, isoforms. The effects of mitochondrially produced NO on bioenergetics were studied in intact cardiomyocytes isolated from dystrophin-deficient (mdx) mice. mdx cardiomyocytes are also deficient in cellular endothelial NOS, but overexpress mtNOS, which allowed us to study the mitochondrial enzyme in intact cells free of its cytosolic counterpart. In these cardiomyocytes, which produce NO beat-to-beat, inhibition of mtNOS increased myocyte shortening by approximately one-fourth. Beat-to-beat NO production and altered shortening by NOS inhibition were not observed in wild-type cells. A plausible mechanism for the reversible NO inhibition of contractility in these cells involves the reaction of NO with cytochrome c oxidase. This suggests a modulatory role for NO in oxidative phosphorylation and, in turn, myocardial contractility.


Asunto(s)
Mitocondrias Cardíacas/enzimología , Óxido Nítrico Sintasa/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio , Distrofina/genética , Distrofina/fisiología , Electroquímica , Complejo IV de Transporte de Electrones/metabolismo , Ratones , Ratones Noqueados , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo I , Superóxidos/metabolismo
12.
Mol Ther ; 4(1): 45-51, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11472105

RESUMEN

Mutations in the gene encoding dystrophin, a large cytoskeletal protein in muscle, lead to Duchenne muscular dystrophy (DMD). Affected individuals often die of respiratory failure resulting primarily from diaphragm muscle degeneration. Here we report a new procedure to transfer the full-length dystrophin cDNA into the diaphragm muscle of Dmd(mdx/mdx) mice, which carry a mutation in the dystrophin gene (Dmd). Significant gene transfer was found after intravenous injection of naked plasmid DNA followed by a brief (eight second) occlusion of blood flow at the vena cava. This is the first demonstration of gene transfer into the diaphragm muscle through systemic administration of naked plasmid DNA. The approach has potential application for treatment of DMD.


Asunto(s)
Diafragma , Distrofina/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Distrofia Muscular Animal/terapia , Plásmidos , Animales , Expresión Génica , Terapia Genética/métodos , Inmunohistoquímica , Inyecciones Intravenosas , Masculino , Ratones , Ratones Endogámicos mdx , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Plásmidos/administración & dosificación , Plásmidos/metabolismo , Factores de Tiempo , Distribución Tisular
13.
Mol Ther ; 3(6): 892-900, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11407903

RESUMEN

Multiple forms of muscular dystrophy are due to the absence of cytoskeletal muscle proteins that normally protect the integrity of muscle cells. The lack of any adequate treatments for these devastating diseases propels research toward the development of strategies for gene delivery to skeletal muscle. High-capacity adenoviral vectors (HC-AdV) devoid of all viral coding sequences have been developed to avoid expression of viral proteins by the gene therapy vector. However, the capsid proteins that are an essential component of the input viral vector and any residual helper virus in the vector preparation could induce an immune response. Furthermore, the therapeutic protein provided by a gene transfer vector presents the potential to induce an immune response in a patient who does not express a normal cellular protein due to genetic mutation. Therefore, we hypothesize that some immune suppression will be required with therapeutic gene delivery designed for the treatment of patients with inherited muscle diseases. In this study, we constructed and rescued three HC-AdVs expressing murine CTLA4Ig, murine CD40Ig, or both. The backbone vector without a gene insert was rescued as a negative control vector. The production of relevant proteins from each vector was determined in vitro. In vivo function of each of the immunosuppressant vectors was assayed by co-injection with an enhanced green fluorescent protein (EGFP)-expressing first-generation adenoviral vector (AdEGFP) into the tibialis anterior muscle of C57BL/10 mice. Higher levels of muscle EGFP expression were observed in animals receiving an immunosuppressant vector. Furthermore, the production of total anti-AdV and anti-EGFP antibodies was reduced in mice treated with each of the three immunosuppressant vectors. A second intramuscular administration of AdEGFP alone 4 weeks after the initial co-injection was successful in all immunosuppressant vector-treated groups, but not in the negative control vector-treated group. All groups had a high antibody response to adenoviral proteins after the second injection of AdEGFP alone, indicating that the initial co-injection did not tolerize against vector capsid antigens.


Asunto(s)
Adenoviridae/genética , Antígenos de Diferenciación/genética , Antígenos CD40/genética , Terapia Genética/métodos , Inmunoconjugados , Músculo Esquelético/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Abatacept , Animales , Antígenos CD , Antígenos de Diferenciación/metabolismo , Western Blotting , Antígenos CD40/metabolismo , Ligando de CD40 , Antígeno CTLA-4 , Distrofina/genética , Citometría de Flujo , Expresión Génica , Vectores Genéticos , Proteínas Fluorescentes Verdes , Inmunoglobulina G/inmunología , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos C57BL , Linfocitos T/inmunología
14.
Muscle Nerve ; 24(6): 787-93, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11360262

RESUMEN

Measurements of muscle strength in clinical trials of Duchenne muscular dystrophy have relied heavily on manual muscle testing (MMT). The high level of intra- and interrater variability of MMT compromises clinical study results. We compared the reliability of 12 clinical evaluators in performing MMT and quantitative muscle testing (QMT) on 12 children with muscular dystrophy. QMT was reliable, with an interclass correlation coefficient (ICC) of >0.9 for biceps and grip strength, and >0.8 for quadriceps strength. Training of both subjects and evaluators was easily accomplished. MMT was not as reliable, and required repeated training of evaluators to bring all groups to an ICC >0.75 for shoulder abduction, elbow and hip flexion, knee extension, and ankle dorsiflexion. We conclude that QMT shows greater reliability and is easier to implement than MMT. Consequently, QMT will be a superior measure of strength for use in pediatric, neuromuscular, multicenter clinical trials.


Asunto(s)
Fuerza de la Mano/fisiología , Músculo Esquelético/fisiopatología , Distrofias Musculares/fisiopatología , Niño , Mano , Humanos , Articulaciones/fisiopatología , Persona de Mediana Edad , Distrofia Muscular de Duchenne/fisiopatología , Variaciones Dependientes del Observador , Reproducibilidad de los Resultados
15.
Curr Neurol Neurosci Rep ; 1(1): 89-96, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11898504

RESUMEN

Gene transfer research for Duchenne muscular dystrophy (DMD) has brought the goal of successful treatment of this devastating, inherited disease closer to being a reality. Although gene therapeutic approaches for DMD patients are not yet in clinical use, recent advances using DMD animal models are encouraging. Progress in vector design, such as high-capacity adenoviral vectors, targeted adenoviral vectors, and heterodimerization of DNA delivered by adeno-associated virus (AAV) vectors have advanced the field considerably. The recent studies into the pharmacologic-induced read-through of stop codons, the increased study of utrophin and its upregulation, and the introduction of point mutation correction using chimeric oligonucleotides have expanded the field, providing new avenues of inquiry.


Asunto(s)
Terapia Genética , Distrofia Muscular de Duchenne/terapia , Antibacterianos/uso terapéutico , Proteínas del Citoesqueleto/uso terapéutico , Vectores Genéticos , Gentamicinas/uso terapéutico , Humanos , Proteínas de la Membrana/uso terapéutico , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/genética , Utrofina
16.
Hum Gene Ther ; 10(10): 1633-40, 1999 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10428208

RESUMEN

Adenoviral vectors (ADVs) are used widely for gene delivery to different tissues including muscle. One particularly promising use for ADVs is in the transfer of the dystrophin gene to the muscle of patients with Duchenne muscular dystrophy (DMD). However, studies in different animal models of DMD suggest that ADVs inefficiently transduce mature skeletal muscle. In this article we test whether AdZ.F(pK7), a genetically modified ADV that expresses a polylysine moiety on the end of the fiber protein, could enhance transduction of muscle cells and circumvent the maturation-dependent loss of muscle infectivity by ADVs. The efficiency of transduction was tested at different levels of muscle maturation. In vitro, AdZ.F(pK7) showed a higher level of transduction at all stages of differentiation including myoblasts, myotubes, and single muscle fibers. In vivo, mature skeletal muscle was transduced fourfold better by AdZ.F(pK7) than by the unmodifled vector (AdZ.F). Together, these observations demonstrate improved ADV transduction of skeletal muscle by modifying ADV tropism, and provide a proof-of-principle that modification of ADVs to target muscle-specific molecules could result in tissue-specific transfer of skeletal muscle tissue as well.


Asunto(s)
Adenovirus Humanos , Proteínas de la Cápside , Cápside/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Polilisina , Animales , Línea Celular , Transformación Celular Viral , Humanos , Masculino , Ratones , Músculo Esquelético/citología
17.
Hum Gene Ther ; 10(3): 365-73, 1999 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-10048389

RESUMEN

Previous studies of the use of adenoviral vectors in animal models of gene therapy have focused on the immune response against transduced cells as the major limiting factor to long-term transgene expression. In this study we eliminated the variable of immunity induced by expression of the transgene in order to investigate vector DNA stability of both first-generation and high-capacity adenoviral vectors after gene transfer to skeletal muscle. Transgene expression from a high-capacity adenoviral vector remained at a high level for at least 20 weeks and was accompanied by persistence of intact vector genomes. In contrast, transgene expression from a first-generation adenoviral vector markedly diminished by 6 weeks after gene transfer and was accompanied by mild and variable inflammatory cell infiltrates. Surprisingly, despite this loss of transgene expression, the first-generation adenoviral vector genomes persisted like the high-capacity adenoviral vector genomes. Therefore, in the absence of immunity to transgene proteins, loss of expression from the first-generation vector was due to inhibition of transgene expression rather than to the elimination of vector-containing cells. DNA stability and persistent expression of the high-capacity adenoviral vector supports the potential of this vector for clinical applications of muscle gene transfer.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos , Músculo Esquelético/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Citomegalovirus/genética , Distrofina/genética , Eosina Amarillenta-(YS)/análisis , Femenino , Expresión Génica , Terapia Genética/métodos , Hematoxilina/análisis , Macrófagos/inmunología , Masculino , Ratones , Ratones Transgénicos , Músculos/química , Secuencias Repetidas Terminales/genética , Factores de Tiempo , beta-Galactosidasa/química
18.
Gene Ther ; 5(1): 19-30, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9536261

RESUMEN

Duchenne muscular dystrophy (DMD) is an X-linked recessive muscle disease characterized by a lack of dystrophin expression. Myoblast transplantation and gene therapy have the potential of restoring dystrophin, thus decreasing the muscle weakness associated with this disease. In this study we present data on the myoblast mediated ex vivo gene transfer of full-length dystrophin to mdx (dystrophin deficient) mouse muscle as a model for autologous myoblast transfer. Both isogenic primary mdx myoblasts and an immortalized mdx cell line were transduced with an adenoviral vector that has all viral coding sequences deleted and encodes beta-galactosidase and full-length dystrophin. Subsequently, these transduced myoblasts were injected into dystrophic mdx muscle, where the injected cells restored dystrophin, as well as dystrophin-associated proteins. A greater amount of dystrophin replacement occurred in mdx muscle following transplantation of mdx myoblasts isolated from a transgenic mouse overexpressing dystrophin suggesting that engineering autologous myoblasts to express high amounts of dystrophin might be beneficial. The ex vivo approach possesses attributes that make it useful for gene transfer to skeletal muscle including: (1) creating a reservoir of myoblasts capable of regenerating and restoring dystrophin to dystrophic muscle; and (2) achieving a higher level of gene transfer to dystrophic muscle compared with adenovirus-mediated direct gene delivery. However, as observed in direct gene transfer studies, the ex vivo approach also triggers a cellular immune response which limits the duration of trans-gene expression.


Asunto(s)
Adenoviridae , Distrofina/genética , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Distrofia Muscular Animal/terapia , Animales , Células Cultivadas , Expresión Génica , Ratones , Ratones Endogámicos mdx , Ratones Transgénicos , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Músculo Esquelético/trasplante , beta-Galactosidasa/genética
19.
Hum Gene Ther ; 8(4): 371-80, 1997 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-9054512

RESUMEN

The mechanisms causing age-dependent loss of muscle fiber infectivity observed in vivo for both adenoviral (Ad) and herpes simplex virus type 1 (HSV-1) gene delivery vectors remain poorly understood. Here we investigate the possible bases for this phenomenon using the novel application of enzymatically isolated, viable, single muscle fibers. We show that maturation-dependent loss of fiber infectivity is recapitulated in single fibers, and, thus, is not solely due to host immune response. Using localized irradiation of muscle in vivo, we show data suggesting that Ad infectivity of differentiated myofibers depends, at least in part, on myoblasts to mediate fiber transduction. On the other hand, infection of single fibers by HSV-1 is not affected by irradiation. Using confocal microscopy, we show that the basal lamina of myogenic cells efficiently infected by HSV-1 is structurally less organized than that of fibers resistant to infection by HSV-1. As well, we show that single myofibers isolated from adult, basal lamina-defective mice (merosin-deficient, dy/dy) are at least 10-fold more susceptible to infection by HSV-1 than are myofibers isolated from control mice. Together, these observations support the hypothesis that the basal lamina acts as a physical barrier to HSV-1 infection of mature muscle.


Asunto(s)
Adenoviridae/genética , Envejecimiento , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos , Herpesvirus Humano 1/genética , Fibras Musculares Esqueléticas/virología , Músculo Esquelético/virología , Animales , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Fibras Musculares Esqueléticas/efectos de la radiación , Fibras Musculares Esqueléticas/ultraestructura , Músculo Esquelético/efectos de la radiación , Músculo Esquelético/ultraestructura
20.
Proc Natl Acad Sci U S A ; 94(5): 1645-50, 1997 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-9050832

RESUMEN

Genetic correction of inherited muscle diseases, such as Duchenne muscular dystrophy, will require long term expression of the recombinant protein following gene transfer. We have shown previously that a new adenoviral vector that lacks all viral genes expressed both full-length dystrophin and beta-galactosidase in mdx (dystrophin-deficient) mouse muscle. We observed a significant histologic improvement of vector-transduced mdx muscle before the eventual loss of vector-encoded transgene expression. In this study, we investigated whether an immunological response against vector-encoded beta-galactosidase contributed to the loss of vector expression and affected vector persistence in muscle. Intramuscular vector injection in control normal mice resulted in an early and complete loss of beta-galactosidase expression accompanied by predominantly CD4+ and CD8+ lymphocytic infiltration and a significant loss of vector DNA. In contrast, intramuscular vector injection in lacZ transgenic mice resulted in persistent expression of beta-galactosidase for at least 84 days with no evidence of inflammation or significant loss of vector DNA. Our studies demonstrate that, in the absence of an immune response induced by beta-galactosidase expression, an adenoviral vector lacking all viral genes is stably maintained in muscle.


Asunto(s)
Adenoviridae/genética , Técnicas de Transferencia de Gen , Genes Virales/genética , Vectores Genéticos/genética , Distrofia Muscular Animal/genética , Linfocitos T/inmunología , Adenoviridae/metabolismo , Animales , ADN Viral/metabolismo , Distrofina/genética , Distrofina/metabolismo , Femenino , Dosificación de Gen , Regulación de la Expresión Génica/genética , Terapia Genética , Humanos , Inyecciones Intramusculares , Masculino , Ratones , Ratones Transgénicos , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/terapia , Transgenes/genética , beta-Galactosidasa/genética , beta-Galactosidasa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA