Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Hum Gene Ther ; 29(7): 737-748, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29433343

RESUMO

Heart disease is a major health threat for Duchenne/Becker muscular dystrophy patients and carriers. Expression of a 6-8 kb mini-dystrophin gene in the heart holds promise to change the disease course dramatically. However, the mini-dystrophin gene cannot be easily studied with adeno-associated virus (AAV) gene delivery because the size of the minigene exceeds AAV packaging capacity. Cardiac protection of the ΔH2-R19 minigene was previously studied using the cardiac-specific transgenic approach. Although this minigene fully normalized skeletal muscle force, it only partially corrected electrocardiogram and heart hemodynamics in dystrophin-null mdx mice that had moderate cardiomyopathy. This study evaluated the ΔH2-R15 minigene using the same transgenic approach in mdx mice that had more severe cardiomyopathy. In contrast to the ΔH2-R19 minigene, the ΔH2-R15 minigene carries dystrophin spectrin-like repeats 16 to 19 (R16-19), a region that has been suggested to protect the heart in clinical studies. Cardiac expression of the ΔH2-R15 minigene normalized all aberrant electrocardiogram changes and improved hemodynamics. Importantly, it corrected the end-diastolic volume, an important diastolic parameter not rescued by ΔH2-R19 mini-dystrophin. It is concluded that that ΔH2-R15 mini-dystrophin is a superior candidate gene for heart protection. This finding has important implications in the design of the mini/micro-dystrophin gene for Duchenne cardiomyopathy therapy.


Assuntos
Cardiomiopatia Dilatada/terapia , Distrofina/genética , Terapia Genética , Miocárdio/metabolismo , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/fisiopatologia , Modelos Animais de Doenças , Distrofina/administração & dosagem , Distrofina/biossíntese , Eletrocardiografia , Regulação da Expressão Gênica/genética , Vetores Genéticos/genética , Vetores Genéticos/uso terapêutico , Humanos , Camundongos , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatologia , Distrofia Muscular Animal/terapia , Miocárdio/patologia , Volume Sistólico/genética
2.
Mol Ther ; 18(6): 1210-7, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20407428

RESUMO

Antisense oligonucleotides (AONs) are being developed as RNA therapeutic molecules for Duchenne muscular dystrophy. For oligonucleotides with the 2'-O-methyl-phosphorothioate (2OMePS) RNA chemistry, proof of concept has been obtained in patient-specific muscle cell cultures, the mouse and dog disease models, and recently by local administration in Duchenne patients. To further explore the pharmacokinetic (PK)/pharmacodynamic (PD) properties of this chemical class of oligonucleotides, we performed a series of preclinical studies in mice. The results demonstrate that the levels of oligonucleotides in dystrophin-deficient muscle fibers are much higher than in healthy fibers, leading to higher exon-skipping levels. Oligonucleotide levels and half-life differed for specific muscle groups, with heart muscle showing the lowest levels but longest half-life (approximately 46 days). Intravenous (i.v.), subcutaneous (s.c.), and intraperitoneal (i.p.) delivery methods were directly compared. For each method, exon-skipping and novel dystrophin expression were observed in all muscles, including arrector pili smooth muscle in skin biopsies. After i.v. administration, the oligonucleotide peak levels in plasma, liver, and kidney were higher than after s.c. or i.p. injections. However, as the bioavailability was similar, and the levels of oligonucleotide, exon-skipping, and dystrophin steadily accumulated overtime after s.c. administration, we selected this patient-convenient delivery method for future clinical study protocols.


Assuntos
Oligonucleotídeos Fosforotioatos/farmacologia , RNA Antissenso/farmacologia , Animais , Western Blotting , Modelos Animais de Doenças , Distrofina/administração & dosagem , Imunofluorescência , Camundongos , Camundongos Endogâmicos mdx , Oligonucleotídeos Fosforotioatos/farmacocinética , RNA Antissenso/farmacocinética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
3.
Arch Phys Med Rehabil ; 90(1): 66-73, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19154831

RESUMO

OBJECTIVES: To evaluate the effect of functional overloading on the transplantation of muscle derived stem cells (MDSCs) into dystrophic muscle and the ability of transplanted cells to increase dystrophic muscle's ability to resist overloading-induced weakness. DESIGN: Cross-sectional. SETTING: Laboratory. ANIMALS: Male mice (N=10) with a dystrophin gene mutation. INTERVENTIONS: MDSCs were intramuscularly transplanted into the extensor digitorum longus muscle (EDL). Functional overloading of the EDL was performed by surgical ablation of the EDL's synergist. MAIN OUTCOME MEASURES: The total number of dystrophin-positive fibers/cross-section (as a measure of stem cell engraftment), the average number of CD31+ cells (as a measure of capillarity), and in vitro EDL contractile strength. Independent t tests were used to investigate the effect of overloading on engraftment, capillarity, and strength. Paired t tests were used to investigate the effect of MDSC engraftment on strength and capillarity. RESULTS: MDSC transplantation protects dystrophic muscles against overloading-induced weakness (specific twitch force: control 4.5N/cm2+/-2.3; MDSC treated 7.9N/cm2+/-1.4) (P=.02). This improved force production following overloading is concomitant with an increased regeneration by transplanted MDSCs (MDSC: 26.6+/-20.2 dystrophin-positive fibers/cross-section; overloading + MDSC: 170.6+/-130.9 dystrophin-positive fibers/cross-section [P=.03]). Overloading-induced increases in skeletal muscle capillarity is significantly correlated with increased MDSC engraftment (R2=.80, P=.01). CONCLUSIONS: These findings suggest that the functional contribution of transplanted MDSCs may rely on activity-dependent mechanisms, possibly mediated by skeletal muscle vascularity. Rehabilitation modalities may play an important role in the development of stem cell transplantation strategies for the treatment of muscular dystrophy.


Assuntos
Modelos Animais de Doenças , Distrofina/farmacologia , Contração Muscular/fisiologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Distrofia Muscular Animal/terapia , Transplante de Células-Tronco , Animais , Células Cultivadas , Distrofina/administração & dosagem , Distrofina/deficiência , Masculino , Camundongos
4.
Hum Mol Genet ; 12(11): 1287-99, 2003 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-12761044

RESUMO

Dystrophin gene transfer using helper-dependent adenoviruses (HDAd), which are deleted of all viral genes, is a promising option to treat muscles in Duchenne muscular dystrophy. We investigated the benefits of this approach by injecting the tibialis anterior (TA) muscle of neonatal and juvenile (4-6-week-old) dystrophin-deficient (mdx) mice with a fully deleted HDAd (HDCBDysM). This vector encoded two full-length murine dystrophin cDNAs regulated by the powerful cytomegalovirus enhancer/beta-actin promoter. At 10 days post-injection of neonatal muscles, 712 fibers (42% of the total number of TA fibers) were dystrophin-positive (dys+), a value that did not decrease for 6 months (the study duration). In treated juveniles, maximal transduction occurred at 30 days post-injection (414 dys+ fibers, 24% of the total number of TA fibers), but decreased by 51% after 6 months. All studied aspects of the pathology were improved in neonatally treated muscles: the percentage of dys+ fibers with centrally localized myonuclei remained low, localization of the dystrophin associated protein complex was restored at the plasma membrane, muscle hypertrophy was reduced, and maximal force-generating capacity and resistance to contraction-induced injuries were increased. The same pathological aspects were improved in the treated juveniles, except for reduction of muscle hypertrophy and maximal force-generating capacity. We demonstrated a strong humoral response against murine dystrophin in both animal groups, but mild inflammatory response occurred only in the treated juveniles. HDCBDysM is thus one of the most promising and efficient vectors for treating DMD by gene therapy.


Assuntos
Adenoviridae/genética , Distrofina/genética , Distrofina/metabolismo , Músculo Esquelético/fisiologia , Actinas/genética , Fatores Etários , Animais , Animais Recém-Nascidos , Formação de Anticorpos , Distrofina/administração & dosagem , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/efeitos adversos , Vetores Genéticos/genética , Inflamação/imunologia , Injeções Intramusculares , Camundongos , Camundongos Endogâmicos mdx , Fibras Musculares Esqueléticas/patologia , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/fisiopatologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/imunologia , Distrofia Muscular de Duchenne/terapia , Regiões Promotoras Genéticas
5.
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
6.
Mol Ther ; 4(5): 499-507, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11708887

RESUMO

Duchenne muscular dystrophy (DMD) is caused by defects in the dystrophin gene. In young dystrophic mdx mice, immature regenerating myofibers represent the principal substrate for adenovirus vector (AdV)-mediated dystrophin gene transfer. However, in DMD patients immature regenerating myofibers are generally sparse. Such a situation also exists in old mdx mice, which may represent a more realistic model. Therefore, here we have used old mdx mice (of 14- to 17 months of age) to test the hypothesis that one-time administration of a myonecrotic agent can transiently re-establish a population of immature myofibers susceptible to AdV-mediated dystrophin gene transfer. This strategy led to upregulation of the coxsackie/adenovirus attachment receptor by means of induction of regenerating myofibers, significantly augmented AdV-mediated dystrophin gene expression, and enhanced force-generating capacity. In addition, it led to an increased resistance to contraction-induced injury compared with untreated controls. The latter protective effect was positively correlated with the number of dystrophin-expressing myofibers (r=0.83, P<0.05). Accordingly, the risk:benefit ratio associated with the sequential use of forced myofiber regeneration and AdV-mediated dystrophin gene transfer was favorable in old mdx mice despite advanced disease. These findings have implications for the potential applicability of AdV-mediated gene therapy to DMD and other muscle diseases in which immature regenerating myofibers are lacking.


Assuntos
Envelhecimento/fisiologia , Distrofina/genética , Distrofina/uso terapêutico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatologia , Miofibrilas/fisiologia , Regeneração , Adenoviridae/genética , Animais , Western Blotting , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Distrofina/administração & dosagem , Venenos Elapídicos/farmacologia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/genética , Imuno-Histoquímica , Contração Isométrica , Camundongos , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/terapia , Miofibrilas/efeitos dos fármacos , Miofibrilas/genética , Miofibrilas/patologia , Receptores Virais/biossíntese , Receptores Virais/genética , Regeneração/efeitos dos fármacos , Estresse Mecânico , Transgenes/genética , Regulação para Cima/efeitos dos fármacos
7.
Gene Ther ; 6(8): 1406-14, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10467365

RESUMO

Patterns of dystrophin and beta-galactosidase expression were examined in mdx mice after i.m. injections of synthetic microspheres (MF-2) loaded with full-length (pHSADy) or mini-dystrophin gene (pSG5dys) cDNA plasmid constructs or with LacZ marker gene (pCMV-LacZ). A single injection of 25 microg pHSADy into quadriceps femoris muscle resulted in 6.8% of dystrophin positive myofibers (DPM) in a given muscle; 8.4% of DPM in glutaeus muscle and 4.3% of DPM in quadriceps femoris muscle of contralateral limb on day 21 after exposure compared with only 0.6% DPM in intact (non-injected) mdx mice. A high proportion of DPM (17.6% and 10.8%, respectively) was registered in both injected and contralateral muscles after mini- gene cDNA administration. MF-2/dystrophin cDNA particles were detected by FISH analysis in about 60-70% of myofiber nuclei in muscles of injected and contralateral limbs 7 days after application. The presence of human dystrophin cDNA and its products in all skeletal muscles and in different internal organs was proven by PCR and RT-PCR analysis. Patches of beta-galactosidase expression were abundant in injected muscle, and frequent in the contralateral and other skeletal muscles as well as in diaphragm, heart and lungs. High levels of dystrophin cDNA expression, and an efficient distant transfection effect with preferential intranuclei inclusion of MF-2 vehicle, are very encouraging for the development of a new constructive strategy in gene therapy trials of DMD.


Assuntos
Distrofina/genética , Técnicas de Transferência de Genes , Óperon Lac/genética , Fibras Musculares Esqueléticas/fisiologia , Transfecção/genética , Animais , DNA Complementar/administração & dosagem , Distrofina/administração & dosagem , Distrofina/metabolismo , Terapia Genética/métodos , Humanos , Injeções Intramusculares , Camundongos , Camundongos Endogâmicos mdx , Microesferas , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/terapia , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...