Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Skelet Muscle ; 7(1): 27, 2017 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-29241457

RESUMEN

BACKGROUND: Previous studies in patients with limb-girdle muscular dystrophy type 2A (LGMD2A) have suggested that calpain-3 (CAPN3) mutations result in aberrant regeneration in muscle. METHODS: To gain insight into pathogenesis of aberrant muscle regeneration in LGMD2A, we used a paradigm of cardiotoxin (CTX)-induced cycles of muscle necrosis and regeneration in the CAPN3-KO mice to simulate the early features of the dystrophic process in LGMD2A. The temporal evolution of the regeneration process was followed by assessing the oxidative state, size, and the number of metabolic fiber types at 4 and 12 weeks after last CTX injection. Muscles isolated at these time points were further investigated for the key regulators of the pathways involved in various cellular processes such as protein synthesis, cellular energy status, metabolism, and cell stress to include Akt/mTORC1 signaling, mitochondrial biogenesis, and AMPK signaling. TGF-ß and microRNA (miR-1, miR-206, miR-133a) regulation were also assessed. Additional studies included in vitro assays for quantifying fusion index of myoblasts from CAPN3-KO mice and development of an in vivo gene therapy paradigm for restoration of impaired regeneration using the adeno-associated virus vector carrying CAPN3 gene in the muscle. RESULTS: At 4 and 12 weeks after last CTX injection, we found impaired regeneration in CAPN3-KO muscle characterized by excessive numbers of small lobulated fibers belonging to oxidative metabolic type (slow twitch) and increased connective tissue. TGF-ß transcription levels in the regenerating CAPN3-KO muscles were significantly increased along with microRNA dysregulation compared to wild type (WT), and the attenuated radial growth of muscle fibers was accompanied by perturbed Akt/mTORC1 signaling, uncoupled from protein synthesis, through activation of AMPK pathway, thought to be triggered by energy shortage in the CAPN3-KO muscle. This was associated with failure to increase mitochondria content, PGC-1α, and ATP5D transcripts in the regenerating CAPN3-KO muscles compared to WT. In vitro studies showed defective myotube fusion in CAPN3-KO myoblast cultures. Replacement of CAPN3 by gene therapy in vivo increased the fiber size and decreased the number of small oxidative fibers. CONCLUSION: Our findings provide insights into understanding of the impaired radial growth phase of regeneration in calpainopathy.


Asunto(s)
Calpaína/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/fisiología , Biogénesis de Organelos , Regeneración , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Calpaína/genética , Células Cultivadas , Modelos Animales de Enfermedad , Terapia Genética , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/metabolismo , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/fisiopatología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
2.
Hum Gene Ther ; 26(10): 647-56, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26076707

RESUMEN

Duchenne muscular dystrophy (DMD) is caused by mutations in the DMD gene. It is the most common, severe childhood form of muscular dystrophy. We investigated an alternative to dystrophin replacement by overexpressing ITGA7 using adeno-associated virus (AAV) delivery. ITGA7 is a laminin receptor in skeletal muscle that, like the dystrophin-glycoprotein complex, links the extracellular matrix to the internal actin cytoskeleton. ITGA7 is expressed in DMD patients and overexpression does not elicit an immune response to the transgene. We delivered rAAVrh.74.MCK.ITGA7 systemically at 5-7 days of age to the mdx/utrn(-/-) mouse deficient for dystrophin and utrophin, a severe mouse model of DMD. At 8 weeks postinjection, widespread expression of ITGA7 was observed at the sarcolemma of multiple muscle groups following gene transfer. The increased expression of ITGA7 significantly extended longevity and reduced common features of the mdx/utrn(-/-) mouse, including kyphosis. Overexpression of α7 expression protected against loss of force following contraction-induced damage and increased specific force in the diaphragm and EDL muscles 8 weeks after gene transfer. Taken together, these results further support the use of α7 integrin as a potential therapy for DMD.


Asunto(s)
Antígenos CD/genética , Distrofina/genética , Cadenas alfa de Integrinas/genética , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Animales , Antígenos CD/administración & dosificación , Antígenos CD/biosíntesis , Dependovirus , Modelos Animales de Enfermedad , Distrofina/deficiencia , Regulación de la Expresión Génica , Terapia Genética/métodos , Humanos , Cadenas alfa de Integrinas/administración & dosificación , Cadenas alfa de Integrinas/biosíntesis , Ratones , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología
3.
Ann Clin Transl Neurol ; 2(3): 256-70, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25815352

RESUMEN

OBJECTIVE: Dysferlinopathies are a family of untreatable muscle disorders caused by mutations in the dysferlin gene. Lack of dysferlin protein results in progressive dystrophy with chronic muscle fiber loss, inflammation, fat replacement, and fibrosis; leading to deteriorating muscle weakness. The objective of this work is to demonstrate efficient and safe restoration of dysferlin expression following gene therapy treatment. METHODS: Traditional gene therapy is restricted by the packaging capacity limit of adeno-associated virus (AAV), however, use of a dual vector strategy allows for delivery of over-sized genes, including dysferlin. The two vector system (AAV.DYSF.DV) packages the dysferlin cDNA utilizing AAV serotype rh.74 through the use of two discrete vectors defined by a 1 kb region of homology. Delivery of AAV.DYSF.DV via intramuscular and vascular delivery routes in dysferlin deficient mice and nonhuman primates was compared for efficiency and safety. RESULTS: Treated muscles were tested for dysferlin expression, overall muscle histology, and ability to repair following injury. High levels of dysferlin overexpression was shown for all muscle groups treated as well as restoration of functional outcome measures (membrane repair ability and diaphragm specific force) to wild-type levels. In primates, strong dysferlin expression was demonstrated with no safety concerns. INTERPRETATION: Treated muscles showed high levels of dysferlin expression with functional restoration with no evidence of toxicity or immune response providing proof of principle for translation to dysferlinopathy patients.

4.
Mol Ther ; 22(4): 713-24, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24145553

RESUMEN

Overexpression of GALGT2 in skeletal muscle can stimulate the glycosylation of α dystroglycan and the upregulation of normally synaptic dystroglycan-binding proteins, some of which are dystrophin and laminin α2 surrogates known to be therapeutic for several forms of muscular dystrophy. This article describes the vascular delivery of GALGT2 gene therapy in a large animal model, the rhesus macaque. Recombinant adeno-associated virus, rhesus serotype 74 (rAAVrh74), was used to deliver GALGT2 via the femoral artery to the gastrocnemius muscle using an isolated focal limb perfusion method. GALGT2 expression averaged 44 ± 4% of myofibers after treatment in macaques with low preexisting anti-rAAVrh74 serum antibodies, and expression was reduced to 9 ± 4% of myofibers in macaques with high preexisting rAAVrh74 immunity (P < 0.001; n = 12 per group). This was the case regardless of the addition of immunosuppressants, including prednisolone, tacrolimus, and mycophenolate mofetil. GALGT2-treated macaque muscles showed increased glycosylation of α dystroglycan and increased expression of dystrophin and laminin α2 surrogate proteins, including utrophin, plectin1, agrin, and laminin α5. These experiments demonstrate successful transduction of rhesus macaque muscle with rAAVrh74.MCK.GALGT2 after vascular delivery and induction of molecular changes thought to be therapeutic in several forms of muscular dystrophy.


Asunto(s)
Distrofina/biosíntesis , Técnicas de Transferencia de Gen , Terapia Genética , Laminina/biosíntesis , Distrofias Musculares/genética , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Distroglicanos/genética , Distroglicanos/metabolismo , Distrofina/genética , Regulación de la Expresión Génica , Glicosiltransferasas/genética , Laminina/genética , Macaca mulatta/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofias Musculares/patología , Distrofias Musculares/terapia
5.
Hum Mol Genet ; 22(24): 4929-37, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23863459

RESUMEN

Pharmacologic strategies have provided modest improvement in the devastating muscle-wasting disease, Duchenne muscular dystrophy (DMD). Pre-clinical gene therapy studies have shown promise in the mdx mouse model; however, studies conducted after disease onset fall short of fully correcting muscle strength or protecting against contraction-induced injury. Here we examine the treatment effect on muscle physiology in aged dystrophic mice with significant disease pathology by combining two promising therapies: micro-dystrophin gene replacement and muscle enhancement with follistatin, a potent myostatin inhibitor. Individual treatments with micro-dystrophin and follistatin demonstrated marked improvement in mdx mice but were insufficient to fully restore muscle strength and response to injury to wild-type levels. Strikingly, when combined, micro-dystrophin/follistatin treatment restored force generation and conferred resistance to contraction-induced injury in aged mdx mice. Pre-clinical studies with miniature dystrophins have failed to demonstrate full correction of the physiological defects seen in mdx mice. Importantly, the addition of a muscle enhancement strategy with delivery of follistatin in combination with micro-dystrophin gene therapy completely restored resistance to eccentric contraction-induced injury and improved force. Eccentric contraction-induced injury is a pre-clinical parameter relevant to the exercise induced injury that occurs in DMD patients, and herein, we demonstrate compelling evidence for the therapeutic potential of micro-dystrophin/follistatin combinatorial therapy.


Asunto(s)
Distrofina/genética , Folistatina/genética , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Distrofina/metabolismo , Folistatina/metabolismo , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Ratones , Ratones Endogámicos mdx , Contracción Muscular/genética , Fuerza Muscular/genética , Músculo Esquelético/patología , Distrofia Muscular Animal , Distrofia Muscular de Duchenne/terapia
6.
Mol Ther ; 21(3): 520-5, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23319059

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe muscle disease caused by mutations in the DMD gene, with loss of its gene product, dystrophin. Dystrophin helps link integral membrane proteins to the actin cytoskeleton and stabilizes the sarcolemma during muscle activity. We investigated an alternative therapeutic approach to dystrophin replacement by overexpressing human α7 integrin (ITGA7) using adeno-associated virus (AAV) delivery. ITGA7 is a laminin receptor in skeletal and cardiac muscle that links the extracellular matrix (ECM) to the actin skeleton. It is modestly upregulated in DMD muscle and has been proposed to be an important modifier of dystrophic symptoms. We delivered rAAV8.MCK.ITGA7 to the lower limb of mdx mice through isolated limb perfusion (ILP) of the femoral artery. We demonstrated ~50% of fibers in the tibialis anterior (TA) and extensor digitorum longus (EDL) overexpressing α7 integrin at the sarcolemma following AAV gene transfer. The increase in ITGA7 in skeletal muscle significantly protected against loss of force following eccentric contraction-induced injury compared with untreated (contralateral) muscles while specific force following tetanic contraction was unchanged. Reversal of additional dystrophic features included reduced Evans blue dye (EBD) uptake and increased muscle fiber diameter. Taken together, this data shows that rAAV8.MCK.ITGA7 gene transfer stabilizes the sarcolemma potentially preserving mdx muscle from further damage. This therapeutic approach demonstrates promise as a viable treatment for DMD with further implications for other forms of muscular dystrophy.


Asunto(s)
Antígenos CD/genética , Dependovirus/genética , Vectores Genéticos , Cadenas alfa de Integrinas/genética , Distrofia Muscular de Duchenne/terapia , Animales , Antígenos CD/metabolismo , Modelos Animales de Enfermedad , Distrofina/genética , Distrofina/metabolismo , Matriz Extracelular/metabolismo , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Humanos , Cadenas alfa de Integrinas/metabolismo , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatología , Mutación , Sarcolema/genética , Regulación hacia Arriba
7.
PLoS One ; 7(6): e39233, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22720081

RESUMEN

The dysferlinopathies comprise a group of untreatable muscle disorders including limb girdle muscular dystrophy type 2B, Miyoshi myopathy, distal anterior compartment syndrome, and rigid spine syndrome. As with other forms of muscular dystrophy, adeno-associated virus (AAV) gene transfer is a particularly auspicious treatment strategy, however the size of the DYSF cDNA (6.5 kb) negates packaging into traditional AAV serotypes known to express well in muscle (i.e. rAAV1, 2, 6, 8, 9). Potential advantages of a full cDNA versus a mini-gene include: maintaining structural-functional protein domains, evading protein misfolding, and avoiding novel epitopes that could be immunogenic. AAV5 has demonstrated unique plasticity with regards to packaging capacity and recombination of virions containing homologous regions of cDNA inserts has been implicated in the generation of full-length transcripts. Herein we show for the first time in vivo that homologous recombination following AAV5.DYSF gene transfer leads to the production of full length transcript and protein. Moreover, gene transfer of full-length dysferlin protein in dysferlin deficient mice resulted in expression levels sufficient to correct functional deficits in the diaphragm and importantly in skeletal muscle membrane repair. Intravascular regional gene transfer through the femoral artery produced high levels of transduction and enabled targeting of specific muscle groups affected by the dysferlinopathies setting the stage for potential translation to clinical trials. We provide proof of principle that AAV5 mediated delivery of dysferlin is a highly promising strategy for treatment of dysferlinopathies and has far-reaching implications for the therapeutic delivery of other large genes.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Proteínas de la Membrana/genética , Proteínas Musculares/genética , Recombinación Genética , Disferlina , Humanos
8.
Methods Mol Biol ; 709: 287-98, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21194036

RESUMEN

The development of a nonhuman primate (NHP) model for vascular delivery of therapeutic transgenes with adeno-associated viral (AAV) vectors is crucial for successfully treating muscular dystrophies. Current animal models for Duchenne muscular dystrophy (DMD) gene therapy have species limitations related to assessing function, immune response, and distribution of the micro- and minidystrophin transgenes in a clinically relevant manner. In addition, there are many forms of muscular dystrophy for which there are no available disease models. NHPs provide the ideal model to optimize vector delivery across a vascular barrier and provide accurate dose estimates for local or broadly targeted gene therapy studies. The vascular anatomy NHPs more clearly parallels humans providing an appropriate substrate for translational experiments. Here we outline the development of a rhesus macaque isolated focal limb perfusion (IFLP) protocol targeting the vascular bed of the gastrocnemius. This protocol serves as a model with broad implications for other muscle diseases along with the capability of targeting multiple muscle groups. To overcome the partial homogeneity between portions of the human microdystrophin transgene and those of the NHP dystrophin gene, we utilized a FLAG tag for tracking distribution of microdystrophin. We also provide methods for assessing transduction efficiency of microdystrophin.FLAG following the IFLP vascular delivery protocol.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Distrofia Muscular Animal/terapia , Perfusión/métodos , Animales , Capilares , Permeabilidad Capilar , Forma MM de la Creatina-Quinasa/genética , Modelos Animales de Enfermedad , Distrofina/genética , Extremidades , Macaca mulatta , Modelos Animales , Músculo Esquelético , Distrofia Muscular Animal/genética , Oligopéptidos , Péptidos/genética , Regiones Promotoras Genéticas , Transgenes
9.
Mol Ther ; 18(1): 109-17, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19904237

RESUMEN

Animal models for Duchenne muscular dystrophy (DMD) have species limitations related to assessing function, immune response, and distribution of micro- or mini-dystrophins. Nonhuman primates (NHPs) provide the ideal model to optimize vector delivery across a vascular barrier and provide accurate dose estimates for widespread transduction. To address vascular delivery and dosing in rhesus macaques, we have generated a fusion construct that encodes an eight amino-acid FLAG epitope at the C-terminus of micro-dystrophin to facilitate translational studies targeting DMD. Intramuscular (IM) injection of AAV8.MCK.micro-dys.FLAG in the tibialis anterior (TA) of macaques demonstrated robust gene expression, with muscle transduction (50-79%) persisting for up to 5 months. Success by IM injection was followed by targeted vascular delivery studies using a fluoroscopy-guided catheter threaded through the femoral artery. Three months after gene transfer, >80% of muscle fibers showed gene expression in the targeted muscle. No cellular immune response to AAV8 capsid, micro-dystrophin, or the FLAG tag was detected by interferon-gamma (IFN-gamma) enzyme-linked immunosorbent spot (ELISpot) at any time point with either route. In summary, an epitope-tagged micro-dystrophin cassette enhances the ability to evaluate site-specific localization and distribution of gene expression in the NHP in preparation for vascular delivery clinical trials.


Asunto(s)
Distrofina/metabolismo , Inyecciones Intraarteriales/métodos , Inyecciones Intramusculares/métodos , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/terapia , Péptidos/metabolismo , Animales , Western Blotting , Dependovirus/genética , Distrofina/genética , Ensayo de Inmunoadsorción Enzimática , Terapia Genética , Vectores Genéticos/genética , Humanos , Macaca mulatta , Ratones , Ratones Endogámicos C57BL , Oligopéptidos , Péptidos/genética
10.
Cell ; 137(6): 1005-17, 2009 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-19524505

RESUMEN

Therapeutic strategies based on modulation of microRNA (miRNA) activity hold great promise due to the ability of these small RNAs to potently influence cellular behavior. In this study, we investigated the efficacy of a miRNA replacement therapy for liver cancer. We demonstrate that hepatocellular carcinoma (HCC) cells exhibit reduced expression of miR-26a, a miRNA that is normally expressed at high levels in diverse tissues. Expression of this miRNA in liver cancer cells in vitro induces cell-cycle arrest associated with direct targeting of cyclins D2 and E2. Systemic administration of this miRNA in a mouse model of HCC using adeno-associated virus (AAV) results in inhibition of cancer cell proliferation, induction of tumor-specific apoptosis, and dramatic protection from disease progression without toxicity. These findings suggest that delivery of miRNAs that are highly expressed and therefore tolerated in normal tissues but lost in disease cells may provide a general strategy for miRNA replacement therapies.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , MicroARNs/uso terapéutico , Animales , Ciclina D2 , Ciclinas/metabolismo , Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética
11.
Nat Biotechnol ; 27(1): 59-65, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19098898

RESUMEN

Delivery of genes to the brain and spinal cord across the blood-brain barrier (BBB) has not yet been achieved. Here we show that adeno-associated virus (AAV) 9 injected intravenously bypasses the BBB and efficiently targets cells of the central nervous system (CNS). Injection of AAV9-GFP into neonatal mice through the facial vein results in extensive transduction of dorsal root ganglia and motor neurons throughout the spinal cord and widespread transduction of neurons throughout the brain, including the neocortex, hippocampus and cerebellum. In adult mice, tail vein injection of AAV9-GFP leads to robust transduction of astrocytes throughout the entire CNS, with limited neuronal transduction. This approach may enable the development of gene therapies for a range of neurodegenerative diseases, such as spinal muscular atrophy, through targeting of motor neurons, and amyotrophic lateral sclerosis, through targeting of astrocytes. It may also be useful for rapid postnatal genetic manipulations in basic neuroscience studies.


Asunto(s)
Astrocitos/metabolismo , Dependovirus/genética , Vectores Genéticos , Neuronas/metabolismo , Animales , Ganglios Espinales/metabolismo , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/metabolismo , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Atrofia Muscular Espinal/metabolismo , Neocórtex/metabolismo , Transgenes
12.
Am J Physiol Cell Physiol ; 296(3): C476-88, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19109526

RESUMEN

The cytotoxic T cell (CT) GalNAc transferase, or Galgt2, is a UDP-GalNAc:beta1,4-N-acetylgalactosaminyltransferase that is localized to the neuromuscular synapse in adult skeletal muscle, where it creates the synaptic CT carbohydrate antigen {GalNAcbeta1,4[NeuAc(orGc)alpha2, 3]Galbeta1,4GlcNAcbeta-}. Overexpression of Galgt2 in the skeletal muscles of transgenic mice inhibits the development of muscular dystrophy in mdx mice, a model for Duchenne muscular dystrophy. Here, we provide physiological evidence as to how Galgt2 may inhibit the development of muscle pathology in mdx animals. Both Galgt2 transgenic wild-type and mdx skeletal muscles showed a marked improvement in normalized isometric force during repetitive eccentric contractions relative to nontransgenic littermates, even using a paradigm where nontransgenic muscles had force reductions of 95% or more. Muscles from Galgt2 transgenic mice, however, showed a significant decrement in normalized specific force and in hindlimb and forelimb grip strength at some ages. Overexpression of Galgt2 in muscles of young adult mdx mice, where Galgt2 has no effect on muscle size, also caused a significant decrease in force drop during eccentric contractions and increased normalized specific force. A comparison of Galgt2 and microdystrophin overexpression using a therapeutically relevant intravascular gene delivery protocol showed Galgt2 was as effective as microdystrophin at preventing loss of force during eccentric contractions. These experiments provide a mechanism to explain why Galgt2 overexpression inhibits muscular dystrophy in mdx muscles. That overexpression also prevents loss of force in nondystrophic muscles suggests that Galgt2 is a therapeutic target with broad potential applications.


Asunto(s)
Terapia Genética , Glicosiltransferasas/metabolismo , Contracción Isométrica , Fuerza Muscular , Músculo Esquelético/enzimología , Distrofia Muscular de Duchenne/prevención & control , Adenoviridae/genética , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Miembro Anterior , Terapia Genética/métodos , Vectores Genéticos , Glicosiltransferasas/genética , Miembro Posterior , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Transgénicos , Fibras Musculares Esqueléticas/enzimología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/enzimología , Distrofia Muscular de Duchenne/fisiopatología , Regulación hacia Arriba
13.
Sci Transl Med ; 1(6): 6ra15, 2009 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-20368179

RESUMEN

Antagonists of myostatin, a blood-borne negative regulator of muscle growth produced in muscle cells, have shown considerable promise for enhancing muscle mass and strength in rodent studies and could serve as potential therapeutic agents for human muscle diseases. One of the most potent of these agents, follistatin, is both safe and effective in mice, but similar tests have not been performed in nonhuman primates. To assess this important criterion for clinical translation, we tested an alternatively spliced form of human follistatin that affects skeletal muscle but that has only minimal effects on nonmuscle cells. When injected into the quadriceps of cynomolgus macaque monkeys, a follistatin isoform expressed from an adeno-associated virus serotype 1 vector, AAV1-FS344, induced pronounced and durable increases in muscle size and strength. Long-term expression of the transgene did not produce any abnormal changes in the morphology or function of key organs, indicating the safety of gene delivery by intramuscular injection of an AAV1 vector. Our results, together with the findings in mice, suggest that therapy with AAV1-FS344 may improve muscle mass and function in patients with certain degenerative muscle disorders.


Asunto(s)
Folistatina/genética , Técnicas de Transferencia de Gen , Macaca fascicularis/fisiología , Fuerza Muscular/genética , Músculo Esquelético/crecimiento & desarrollo , Empalme Alternativo , Animales , Dependovirus/genética , Vectores Genéticos , Macaca fascicularis/crecimiento & desarrollo
14.
J Transl Med ; 5: 45, 2007 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-17892583

RESUMEN

BACKGROUND: Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder with monogenic mutations setting the stage for successful gene therapy treatment. We have completed a study that directly deals with the following key issues that can be directly adapted to a gene therapy clinical trial using rAAV considering the following criteria: 1) A regional vascular delivery approach that will protect the patient from widespread dissemination of virus; 2) an approach to potentially facilitate safe passage of the virus for efficient skeletal muscle transduction; 3) the use of viral doses to accommodate current limitations imposed by vector production methods; 4) and at the same time, achieve a clinically meaningful outcome by transducing multiple muscles in the lower limb to prolong ambulation. METHODS: The capacity of AAV1, AAV6 or AAV8 to cross the vascular endothelial barrier carrying a micro-dystrophin cDNA was compared under identical conditions with delivery through a catheter placed in the femoral artery of the mdx mouse. Transduction efficiency was assessed by immuno-staining using an antibody (Manex1a) that recognizes the N-terminus of micro-dystrophin. The degree of physiologic correction was assessed by measuring tetanic force and protection from eccentric contraction in the extensor digitorum longus muscle (EDL). The vascular delivery paradigm found successful in the mouse was carried to the non-human primate to test its potential translation to boys with DMD. RESULTS: Regional vascular delivery resulted in transduction by rAAV8.micro-dystrophin reaching 94.5 +/- 0.9 (1 month), 91.3 +/- 3.1 (2 months), and 89.6 +/- 1.6% (3 months). rAAV6.micro-dystrophin treated animals demonstrated 87.7 +/- 6.8 (1 month), 78.9 +/- 7.4 (2 months), and 81.2 +/- 6.2% (3 months) transduction. In striking contrast, rAAV1 demonstrated very low transduction efficiency [0.9 +/- 0.3 (1 month), 2.1 +/- 0.8 (2 months), and 2.1 +/- 0.7% (3 months)] by vascular delivery. Micro-dystrophin delivered by rAAV8 and rAAV6 through the femoral artery significantly improved tetanic force and protected against eccentric contraction. Mouse studies translated to the hindlimb of cynamologous macaques using a similar vascular delivery paradigm. rAAV8 carrying eGFP in doses proportional to the mouse (5 x 1012 vg/kg in mouse vs 2 x 1012 vg/kg in monkey) demonstrated widespread gene expression [medial gastrocnemius - 63.8 +/- 4.9%, lateral gastrocnemius - 66.0 +/- 4.5%, EDL - 80.2 +/- 3.1%, soleus - 86.4 +/- 1.9%, TA - 72.2 +/- 4.0%. CONCLUSION: These studies demonstrate regional vascular gene delivery with AAV serotype(s) in mouse and non-human primate at doses, pressures and volumes applicable for clinical trials in children with DMD.


Asunto(s)
Distrofina/genética , Endotelio Vascular/fisiopatología , Terapia Genética/métodos , Distrofia Muscular de Duchenne/terapia , Animales , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Humanos , Incidencia , Masculino , Ratones , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/epidemiología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatología , Presión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA