Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...