Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Hum Gene Ther Methods ; 24(4): 270-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23895325

RESUMO

The ability to regulate both the timing and specificity of gene expression mediated by viral vectors will be important in maximizing its utility. We describe the development of an adeno-associated virus (AAV)-based vector with tissue-specific gene regulation, using the ARGENT dimerizer-inducible system. This two-vector system based on AAV serotype 9 consists of one vector encoding a combination of reporter genes from which expression is directed by a ubiquitous, inducible promoter and a second vector encoding transcription factor domains under the control of either a heart- or liver-specific promoter, which are activated with a small molecule. Administration of the vectors via either systemic or intrapericardial injection demonstrated that the vector system is capable of mediating gene expression that is tissue specific, regulatable, and reproducible over induction cycles. Somatic gene transfer in vivo is being considered in therapeutic applications, although its most substantial value will be in basic applications such as target validation and development of animal models.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Transfecção/métodos , Animais , Expressão Gênica , Genes Reporter/genética , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Especificidade de Órgãos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
2.
Curr Protoc Microbiol ; Chapter 14: Unit 14D.3, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23408131

RESUMO

Adeno-associated virus (AAV) is a DNA virus with a small (∼4.7 kb) single-stranded genome. It is a naturally replication-defective parvovirus of the dependovirus group. Recombinant AAV (rAAV), for use as a gene transfer vector, is created by replacing the viral rep and cap genes with the transgene of interest along with promoter and polyadenylation sequences. Only the viral inverted terminal repeats (ITRs) are required in cis for replication and packaging during production. The ITRs are also necessary and sufficient for vector genome processing and persistence during transduction. The tissue tropism of the rAAV vector is determined by the AAV capsid. In this unit we will discuss several methods to deliver rAAV in order to transduce cardiac and/or skeletal muscle, including intravenous delivery, intramuscular delivery, isolated limb infusion, intrapericardial injection in neonatal mice, and left ventricular wall injection in adult rats.


Assuntos
Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Músculo Esquelético , Miocárdio , Transdução Genética , Animais , Camundongos , Ratos
3.
Mol Ther ; 20(3): 580-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22146342

RESUMO

Although restoration of dystrophin expression via exon skipping in both cardiac and skeletal muscle has been successfully demonstrated in the mdx mouse, restoration of cardiac dystrophin expression in large animal models of Duchenne muscular dystrophy (DMD) has proven to be a challenge. In large animals, investigators have focused on using intravenous injection of antisense oligonucleotides (AO) to mediate exon skipping. In this study, we sought to optimize restoration of cardiac dystrophin expression in the golden retriever muscular dystrophy (GRMD) model using percutaneous transendocardial delivery of recombinant AAV6 (rAAV6) to deliver a modified U7 small nuclear RNA (snRNA) carrying antisense sequence to target the exon splicing enhancers of exons 6 and 8 and correct the disrupted reading frame. We demonstrate restoration of cardiac dystrophin expression at 13 months confirmed by reverse transcription-PCR (RT-PCR) and immunoblot as well as membrane localization by immunohistochemistry. This was accompanied by improved cardiac function as assessed by cardiac magnetic resonance imaging (MRI). Percutaneous transendocardial delivery of rAAV6 expressing a modified U7 exon skipping construct is a safe, effective method for restoration of dystrophin expression and improvement of cardiac function in the GRMD canine and may be easily translatable to human DMD patients.


Assuntos
Processamento Alternativo , Dependovirus/genética , Distrofina/genética , Vetores Genéticos/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Animais , Linhagem Celular , Modelos Animais de Doenças , Cães , Distrofina/metabolismo , Ecocardiografia , Éxons , Fibrose , Expressão Gênica , Ordem dos Genes , Técnicas de Transferência de Genes , Vetores Genéticos/farmacocinética , Genoma Viral , Humanos , Imageamento por Ressonância Magnética , Distrofia Muscular de Duchenne/diagnóstico , Miocárdio/patologia , RNA Mensageiro/metabolismo
4.
Methods Mol Biol ; 807: 219-37, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22034032

RESUMO

Cardiac gene transfer may serve as a novel therapeutic approach in the treatment of heart disease. For it to reach its full potential, methods for highly efficient cardiac gene transfer must be available to investigators so that informative preclinical data can be collected and evaluated. We have recently optimized AAV-mediated cardiac gene transfer protocols in both the mouse and rat. In the mouse, we have developed a procedure for intrapericardial delivery of vector in the neonate and successfully applied intravenous injections in adult animals. In the rat, we have developed a procedure for direct injection of vector into the myocardium in adults and established a protocol for vector delivery into the left ventricular anterior wall by ultrasound-targeted destruction of microbubbles loaded with AAV. Each protocol can be used to achieve safe and efficient cardiac gene transfer in the model of choice.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Miocárdio/metabolismo , Miocárdio/patologia , Animais , Animais Recém-Nascidos , Cardiopatias/genética , Cardiopatias/metabolismo , Cardiopatias/terapia , Camundongos , Microbolhas , Ratos
5.
PLoS One ; 6(9): e23818, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21931616

RESUMO

BACKGROUND: Myostatin is a negative regulator of skeletal muscle mass whose activity is upregulated in adult heart failure (HF); however, its role in congenital heart disease (CHD) is unknown. METHODS: We studied myostatin and IGF-1 expression via Western blot in cardiac tissue at varying degrees of myocardial dysfunction and after biventricular support in CHD by collecting myocardial biopsies from four patient cohorts: A) adult subjects with no known cardiopulmonary disease (left ventricle, LV), (Adult Normal), (n = 5); B) pediatric subjects undergoing congenital cardiac surgery with normal RV size and function (right ventricular outflow tract, RVOT), (n = 3); C) pediatric subjects with worsening but hemodynamically stable LV failure [LV and right ventricle (LV, RV,)] with biopsy collected at the time of orthotopic heart transplant (OHT), (n = 7); and D) pediatric subjects with decompensated bi-ventricular failure on BiVAD support with biopsy collected at OHT (LV, RV, BiVAD), (n = 3). RESULTS: The duration of HF was longest in OHT patients compared to BIVAD. The duration of BiVAD support was 4.3±1.9 days. Myostatin expression was significantly increased in LV-OHT compared to RV-OHT and RVOT, and was increased more than double in decompensated biventricular HF (BiVAD) compared to both OHT and RVOT. An increased myostatin/IGF-1 ratio was associated with ventricular dysfunction. CONCLUSIONS: Myostatin expression in increased in CHD, and the myostatin/IGF-1 ratio increases as ventricular function deteriorates. Future investigation is necessary to determine if restoration of the physiologic myostatin/IGF-1 ratio has therapeutic potential in HF.


Assuntos
Cardiopatias/congênito , Cardiopatias/metabolismo , Coração Auxiliar , Fenômenos Mecânicos , Miostatina/metabolismo , Adulto , Criança , Feminino , Regulação da Expressão Gênica , Cardiopatias/complicações , Cardiopatias/cirurgia , Humanos , Lactente , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Fatores de Regulação Miogênica/metabolismo , Disfunção Ventricular Esquerda/complicações
6.
PLoS One ; 6(6): e20856, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21731628

RESUMO

Duchenne muscular dystrophy (DMD) is a degenerative disorder affecting skeletal and cardiac muscle for which there is no effective therapy. Angiotension receptor blockade (ARB) has excellent therapeutic potential in DMD based on recent data demonstrating attenuation of skeletal muscle disease progression during 6-9 months of therapy in the mdx mouse model of DMD. Since cardiac-related death is major cause of mortality in DMD, it is important to evaluate the effect of any novel treatment on the heart. Therefore, we evaluated the long-term impact of ARB on both the skeletal muscle and cardiac phenotype of the mdx mouse. Mdx mice received either losartan (0.6 g/L) (n = 8) or standard drinking water (n = 9) for two years, after which echocardiography was performed to assess cardiac function. Skeletal muscle weight, morphology, and function were assessed. Fibrosis was evaluated in the diaphragm and heart by Trichrome stain and by determination of tissue hydroxyproline content. By the study endpoint, 88% of treated mice were alive compared to only 44% of untreated (p = 0.05). No difference in skeletal muscle morphology, function, or fibrosis was noted in losartan-treated animals. Cardiac function was significantly preserved with losartan treatment, with a trend towards reduction in cardiac fibrosis. We saw no impact on the skeletal muscle disease progression, suggesting that other pathways that trigger fibrosis dominate over angiotensin II in skeletal muscle long term, unlike the situation in the heart. Our study suggests that ARB may be an important prophylactic treatment for DMD-associated cardiomyopathy, but will not impact skeletal muscle disease.


Assuntos
Coração/fisiopatologia , Losartan/administração & dosagem , Losartan/uso terapêutico , Músculo Esquelético/fisiopatologia , Distrofia Muscular Animal/mortalidade , Distrofia Muscular Animal/fisiopatologia , Animais , Fibrose , Coração/efeitos dos fármacos , Sistema de Condução Cardíaco/diagnóstico por imagem , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/metabolismo , Sistema de Condução Cardíaco/fisiopatologia , Testes de Função Cardíaca , Losartan/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Distrofia Muscular Animal/tratamento farmacológico , Miocárdio/metabolismo , Miocárdio/patologia , Análise de Sobrevida , Ultrassonografia
7.
Hum Gene Ther ; 22(12): 1499-509, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21787232

RESUMO

Duchenne muscular dystrophy (DMD) is a lethal, X-linked recessive disease affecting 1 in 3,500 newborn boys for which there is no effective treatment or cure. One novel strategy that has therapeutic potential for DMD is inhibition of myostatin, a negative regulator of skeletal muscle mass that may also promote fibrosis. Therefore, our goal in this study was to evaluate systemic myostatin inhibition in the golden retriever model of DMD (GRMD). GRMD canines underwent liver-directed gene transfer of a self-complementary adeno-associated virus type 8 vector designed to express a secreted dominant-negative myostatin peptide (n = 4) and were compared with age-matched, untreated GRMD controls (n = 3). Dogs were followed with serial magnetic resonance imaging (MRI) for 13 months to assess cross-sectional area and volume of skeletal muscle, then euthanized so that tissue could be harvested for morphological and histological analysis. We found that systemic myostatin inhibition resulted in increased muscle mass in GRMD dogs as assessed by MRI and confirmed at tissue harvest. We also found that hypertrophy of type IIA fibers was largely responsible for the increased muscle mass and that reductions in serum creatine kinase and muscle fibrosis were associated with long-term myostatin inhibition in GRMD. This is the first report describing the effects of long-term, systemic myostatin inhibition in a large-animal model of DMD, and we believe that the simple and effective nature of our liver-directed gene-transfer strategy makes it an ideal candidate for evaluation as a novel therapeutic approach for DMD patients.


Assuntos
Dependovirus/genética , Vetores Genéticos/uso terapêutico , Fígado/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/terapia , Miostatina/antagonistas & inibidores , Animais , Western Blotting , Creatina Quinase/genética , Creatina Quinase/metabolismo , Cães , Fibrose/metabolismo , Fibrose/patologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Fígado/patologia , Imageamento por Ressonância Magnética , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular Animal/patologia , Miostatina/genética , Miostatina/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo
8.
J Vet Cardiol ; 13(2): 131-40, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21640678

RESUMO

Gene therapy is a procedure resulting in the transfer of a gene(s) into an individual's cells to treat a disease, which is designed to produce a protein or functional RNA (the gene product). Although most current gene therapy clinical trials focus on cancer and inherited diseases, multiple studies have evaluated the efficacy of gene therapy to abrogate various forms of heart disease. Indeed, human clinical trials are currently underway. One goal of gene transfer may be to express a functional gene when the endogenous gene is inactive. Alternatively, complex diseases such as end stage heart failure are characterized by a number of abnormalities at the cellular level, many of which can be targeted using gene delivery to alter myocardial protein levels. This review will discuss issues related to gene vector systems, gene delivery strategies and two cardiovascular diseases in dogs successfully treated with therapeutic gene delivery.


Assuntos
Doenças Cardiovasculares/veterinária , Doenças do Gato/terapia , Doenças do Cão/terapia , Técnicas de Transferência de Genes/veterinária , Terapia Genética/veterinária , Animais , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/terapia , Doenças do Gato/genética , Gatos , Doenças do Cão/genética , Cães , Terapia Genética/métodos
9.
Hum Gene Ther ; 22(8): 969-77, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21542669

RESUMO

Derangements in calcium cycling have been described in failing hearts, and preclinical studies have suggested that therapies aimed at correcting this defect can lead to improvements in cardiac function and survival. One strategy to improve calcium cycling would be to inhibit phospholamban (PLB), the negative regulator of SERCA2a that is upregulated in failing hearts. The goal of this study was to evaluate the safety and efficacy of using adeno-associated virus (AAV)-mediated cardiac gene transfer of short hairpin RNA (shRNA) to knock down expression of PLB. Six dogs were treated with self-complementary AAV serotype 6 (scAAV6) expressing shRNA against PLB. Three control dogs were treated with empty AAV6 capsid, and two control dogs were treated with scAAV6 expressing dominant negative PLB. Vector was delivered via a percutaneously inserted cardiac injection catheter. PLB mRNA and protein expression were analyzed in three of six shRNA dogs between days 16 and 26. The other three shRNA dogs and five control dogs were monitored long-term to assess cardiac safety. PLB mRNA was reduced 16-fold, and PLB protein was reduced 5-fold, with treatment. Serum troponin elevation and depressed cardiac function were observed in the shRNA group only at 4 weeks. An enzyme-linked immunospot assay failed to detect any T cells reactive to AAV6 capsid in peripheral blood mononuclear cells, heart, or spleen. Microarray analysis revealed alterations in cardiac expression of several microRNAs with shRNA treatment. AAV6-mediated cardiac gene transfer of shRNA effectively knocks down PLB expression but is associated with severe cardiac toxicity. Toxicity may result from dysregulation of endogenous microRNA pathways.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Técnicas de Transferência de Genes , RNA Interferente Pequeno/genética , Animais , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteínas de Ligação ao Cálcio/metabolismo , Dependovirus/genética , Cães , Expressão Gênica , Técnicas de Silenciamento de Genes , Técnicas de Transferência de Genes/efeitos adversos , Vetores Genéticos , Monócitos/citologia , Monócitos/metabolismo , Miócitos Cardíacos/metabolismo , RNA Mensageiro/metabolismo , Troponina/sangue
10.
Hum Gene Ther ; 22(8): 979-84, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21563985

RESUMO

Heart disease is the leading cause of morbidity and mortality, and cardiac gene transfer has potential as a novel therapeutic approach. We previously demonstrated safe and efficient gene transfer to the canine heart using a percutaneous transendocardial injection procedure to deliver self-complementary (sc) adeno-associated virus 6 (AAV6) vector. In the present study, we proceed with our vertical translation study to evaluate cardiac gene transfer in nonhuman primates (NHPs). We screened approximately 30 adult male rhesus macaques for the presence of neutralizing antibodies against AAV6, AAV8, and AAV9, and then selected seven monkeys whose antibody titers against these three serotypes were lower than 1/5. The animals were then randomized to receive either scAAV6 (n=3), scAAV8 (n=1), or scAAV9 (n=3) vector expressing the enhanced green fluorescent protein (EGFP) reporter gene at a dose of 5.4×10(12) genome copies/kg, which was administered according to a modified version of our previously developed transendocardial injection procedure. One animal treated with scAAV6 died secondary to esophageal intubation. The remaining animals were euthanized 7 days after gene transfer, at which time tissue was collected for analysis of EGFP expression, histopathology, and biodistribution of the vector genome. We found that (i) transendocardial delivery of AAV is safe in the NHP, (ii) AAV6 and AAV8 provide efficient cardiac gene transfer at similar levels and are superior to AAV9, and (iii) AAV6 is more cardiac-specific than AAV8 and AAV9. The results of this NHP study may help guide the development AAV vectors for the treatment of cardiovascular disease in humans.


Assuntos
Dependovirus/genética , Miocárdio/metabolismo , Animais , Técnicas de Transferência de Genes , Genes Reporter , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Macaca mulatta , Masculino , Distribuição Aleatória
11.
Methods Mol Biol ; 709: 369-78, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21194041

RESUMO

Achieving efficient cardiac gene transfer in a large animal model has proven to be technically challenging. Prior strategies have employed cardio-pulmonary bypass or dual catheterization with the aid of vasodilators to deliver vectors, such as adenovirus, adeno-associated virus (AAV) or plasmid DNA. While single-stranded AAV vectors have shown the greatest promise, they suffer from delayed expression, which might be circumvented by using self-complementary vectors. We have recently optimized a cardiac gene transfer protocol in the canine using a percutaneous transendocardial injection catheter to deliver an AAV vector under fluoroscopic guidance. Percutaneous transendocardial injection of self-complementary AAV (scAAV)-6 is a safe, effective method for achieving efficient cardiac gene transfer to approximately 60% of the myocardium.


Assuntos
Dependovirus/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos , Miocárdio , Animais , Cateterismo Cardíaco , Procedimentos Cirúrgicos Cardíacos , Cães , Injeções
13.
Muscle Nerve ; 42(5): 722-30, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20730876

RESUMO

Modulation of transforming growth factor-ß (TGF-ß) signaling to promote muscle growth holds tremendous promise for the muscular dystrophies and other disorders involving the loss of functional muscle mass. Previous studies have focused on the TGF-ß family member myostatin and demonstrated that inhibition of myostatin leads to muscle growth in normal and dystrophic mice. We describe a unique method of systemic inhibition of activin IIB receptor signaling via adeno-associated virus (AAV)-mediated gene transfer of a soluble form of the extracellular domain of the activin IIB receptor to the liver. Treatment of mdx mice with activin IIB receptor blockade led to increased skeletal muscle mass, increased force production in the extensor digitorum longus (EDL), and reduced serum creatine kinase. No effect on heart mass or function was observed. Our results indicate that activin IIB receptor blockade represents a novel and effective therapeutic strategy for the muscular dystrophies.


Assuntos
Receptores de Activinas Tipo II/antagonistas & inibidores , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/patologia , Animais , Western Blotting , Clonagem Molecular , Creatina/sangue , Creatina Quinase/sangue , Dependovirus/genética , Ecocardiografia , Testes de Função Cardíaca , Camundongos , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/fisiopatologia , Distrofia Muscular de Duchenne/fisiopatologia , Miostatina/fisiologia , Tamanho do Órgão/efeitos dos fármacos , Transdução de Sinais , Fator de Crescimento Transformador beta/fisiologia
14.
Vet Clin North Am Small Anim Pract ; 40(4): 717-24, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20610021

RESUMO

Therapeutic gene transfer holds promise as a way to treat dilated cardiomyopathy from any underlying cause because the approach attempts to address metabolic disturbances that occur at the molecular level of the failing heart. Calcium-handling abnormalities and increased rates of apoptosis are abnormalities that occur in many types of heart disease, and gene therapies that target these metabolic defects have proven to be beneficial in numerous rodent models of heart disease. The authors are currently evaluating this approach to treat canine idiopathic dilated cardiomyopathy.


Assuntos
Cardiomiopatia Dilatada/veterinária , Doenças do Cão/terapia , Técnicas de Transferência de Genes/veterinária , Terapia Genética/veterinária , Animais , Cálcio/metabolismo , Cardiomiopatia Dilatada/terapia , Cães , Feminino , Masculino
15.
PLoS One ; 5(4): e10230, 2010 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-20419100

RESUMO

Myostatin is well established as a negative regulator of skeletal muscle growth, but its role in the heart is controversial. Our goal in this study was to characterize myostatin regulation following cardiomyocyte stress and to examine the role of myostatin in the regulation of cardiomyocyte size. Neonatal cardiomyocytes were cultured and stressed with phenylephrine. Adenovirus was used to overexpress myostatin or dominant negative myostatin in culture. Adeno-associated virus was used to overexpress myostatin or dominant negative myostatin in mice. Myostatin is upregulated following cardiomyocyte stress in an Erk-dependent manner that is associated with increased nuclear translocation and DNA binding activity of MEF-2. Myostatin overexpression leads to decreased and myostatin inhibition to increased cardiac growth both in vitro and in vivo due to modulation of Akt and NFAT3 pathways. Myostatin is a negative regulator of cardiac growth, and further studies are warranted to investigate the role of myostatin in the healthy and failing heart.


Assuntos
Miostatina/genética , Transporte Ativo do Núcleo Celular , Animais , Animais Recém-Nascidos , Proliferação de Células , Tamanho Celular , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular , Fatores de Transcrição MEF2 , Camundongos , Miócitos Cardíacos/citologia , Fatores de Regulação Miogênica/metabolismo , Fatores de Transcrição NFATC/metabolismo , Fenilefrina , Proteínas Proto-Oncogênicas c-akt/metabolismo , Estresse Fisiológico , Regulação para Cima/genética
16.
Eur J Heart Fail ; 12(5): 444-53, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20348550

RESUMO

AIMS: Myostatin inhibits myoblast differentiation/proliferation and may play a role in heart failure (HF) and reverse remodelling after left ventricular assist device (LVAD) support. This study sought to characterize myostatin expression and activation in advanced HF before and after LVAD support. METHODS AND RESULTS: Left ventricular tissue pairs were collected at LVAD implantation (core) and at cardiac transplantation/LVAD explantation in patients with advanced ischaemic (ICM-ischaemic cardiomyopathy) and non-ischaemic (DCM-dilated cardiomyopathy) HF. Normal cardiac tissue (control) was obtained from hearts not placed for transplantation. Serum was collected independently from patients with stable DCM HF and from healthy controls. Full-length and cleaved propeptide myostatin levels were quantified by western blot analysis. Dilated cardiomyopathy propeptide levels at core were significantly higher than control and significantly increased after LVAD support. Ischaemic cardiomyopathy propeptide levels were higher than control, but did not change after LVAD support. No changes in full-length levels were seen. Serum myostatin levels were significantly higher in DCM HF patients than in healthy controls. CONCLUSION: This is the first clinical evidence that myostatin activation is increased in HF. Myostatin may affect cardiac hypertrophy and may mediate regression of cellular hypertrophy after mechanical unloading.


Assuntos
Cardiomiopatia Dilatada/fisiopatologia , Insuficiência Cardíaca/fisiopatologia , Coração Auxiliar , Isquemia Miocárdica , Miostatina/biossíntese , Adulto , Análise de Variância , Western Blotting , Proteína Morfogenética Óssea 1 , Cardiomiopatia Dilatada/terapia , Estudos de Casos e Controles , Proliferação de Células , Progressão da Doença , Feminino , Insuficiência Cardíaca/terapia , Humanos , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Músculo Esquelético , Mioblastos , Fosforilação , Estudos Prospectivos , Pressão Propulsora Pulmonar , Fatores de Tempo , Fator de Crescimento Transformador beta/biossíntese
17.
Hum Gene Ther ; 21(6): 750-61, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20113166

RESUMO

Vectors based on several new adeno-associated viral (AAV) serotypes demonstrated strong hepatocyte tropism and transduction efficiency in both small- and large-animal models for liver-directed gene transfer. Efficiency of liver transduction by AAV vectors can be further improved in both murine and nonhuman primate (NHP) animals when the vector genomes are packaged in a self-complementary (sc) format. In an attempt to understand potential molecular mechanism(s) responsible for enhanced transduction efficiency of the sc vector in liver, we performed extensive molecular studies of genome structures of conventional single-stranded (ss) and sc AAV vectors from liver after AAV gene transfer in both mice and NHPs. These included treatment with exonucleases with specific substrate preferences, single-cutter restriction enzyme digestion and polarity-specific hybridization-based vector genome mapping, and bacteriophage phi29 DNA polymerase-mediated and double-stranded circular template-specific rescue of persisted circular genomes. In mouse liver, vector genomes of both genome formats seemed to persist primarily as episomal circular forms, but sc vectors converted into circular forms more rapidly and efficiently. However, the overall differences in vector genome abundance and structure in the liver between ss and sc vectors could not account for the remarkable differences in transduction. Molecular structures of persistent genomes of both ss and sc vectors were significantly more heterogeneous in macaque liver, with noticeable structural rearrangements that warrant further characterizations.


Assuntos
Vetores Genéticos , Fígado/virologia , Animais , DNA de Cadeia Simples/genética , Genoma , Hepatócitos/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pongo , Primatas/genética
18.
PLoS One ; 5(2): e9176, 2010 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-20161803

RESUMO

BACKGROUND: Myostatin inhibition is a promising therapeutic strategy to maintain muscle mass in a variety of disorders, including the muscular dystrophies, cachexia, and sarcopenia. Previously described approaches to blocking myostatin signaling include injection delivery of inhibitory propeptide domain or neutralizing antibodies. METHODOLOGY/PRINCIPAL FINDINGS: Here we describe a unique method of myostatin inhibition utilizing recombinant adeno-associated virus to overexpress a secretable dominant negative myostatin exclusively in the liver of mice. Systemic myostatin inhibition led to increased skeletal muscle mass and strength in control C57 Bl/6 mice and in the dystrophin-deficient mdx model of Duchenne muscular dystrophy. The mdx soleus, a mouse muscle more representative of human fiber type composition, demonstrated the most profound improvement in force production and a shift toward faster myosin-heavy chain isoforms. Unexpectedly, the 11-month-old mdx diaphragm was not rescued by long-term myostatin inhibition. Further, mdx mice treated for 11 months exhibited cardiac hypertrophy and impaired function in an inhibitor dose-dependent manner. CONCLUSIONS/SIGNIFICANCE: Liver-targeted gene transfer of a myostatin inhibitor is a valuable tool for preclinical investigation of myostatin blockade and provides novel insights into the long-term effects and shortcomings of myostatin inhibition on striated muscle.


Assuntos
Fígado/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/genética , Miostatina/genética , Animais , Animais Recém-Nascidos , Western Blotting , Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Distrofia Muscular Animal/patologia , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/terapia , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação , Miostatina/metabolismo , Fatores de Tempo
19.
ILAR J ; 50(2): 199-205, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19293462

RESUMO

Several naturally occurring animal models for human genetic heart diseases offer an excellent opportunity to evaluate potential novel therapies, including gene therapy. Some of these diseases--especially those that result in a structural defect during development (e.g., patent ductus arteriosus, pulmonic stenosis)--would likely be difficult to treat with a therapeutic gene transfer approach. However, the ability to transduce a significant proportion of the myocardial cells should make the various forms of inherited cardiomyopathy amenable to a therapeutic gene transfer approach. Adeno-associated virus may be the ideal vector for cardiac gene therapy since its low immunogenicity allows for stable transgene expression, a crucial factor when considering treatment of a chronic disease. Cardiomyopathies are a major cause of morbidity and mortality in both children and adults, and large animal models are available for the major forms of inherited cardiomyopathy (dilated cardiomyopathy, hypertrophic cardiomyopathy, and arrhythmogenic right ventricular cardiomyopathy). One of these animal models, juvenile dilated cardiomyopathy of Portuguese water dogs, offers an effective means to assess the efficacy of therapeutic gene transfer to alter the course of cardiomyopathy and heart failure. Correction of the abnormal metabolic processes that occur with heart failure (e.g., calcium metabolism, apoptosis) could normalize diseased myocardial function. Gene therapy may offer a promising new approach for the treatment of cardiac disease in both veterinary and human clinical settings.


Assuntos
Doenças Cardiovasculares/genética , Doenças Cardiovasculares/terapia , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Terapia Genética/métodos , Animais , Cálcio/metabolismo , Cães , Vetores Genéticos
20.
Exp Cell Res ; 315(5): 748-59, 2009 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-19166831

RESUMO

The Z-line in each striated muscle has a precisely defined width that corresponds to muscle fiber type, and it can enlarge several fold in nemaline myopathy. To explore the mechanism(s) underlying Z-line width and structure maintenance, a series of sarcomeric-alpha-actinin mutants tagged with myc-epitope was transfected into cultured chick myotubes. By double-staining transfected myotubes with myc and myofibrillar protein antibodies, we found that alpha-actinin mutants with deletion of the region from the beginning of the fourth spectrin repeat to the start of the EF-hands resulted in expansion of Z-line width, often displayed a doublet staining pattern, and resulted in formation of nemaline-like bodies in older myotubes under fluorescence microscope. Yeast-two hybridization analysis demonstrated that this region was involved in vinculin binding, and for vinculin to bind alpha-actinin, residues 1-116 and 258-323 were required. Hence, we have defined a critical region of s-alpha-actinin that affects the width and integrity of the Z-line. This region is at least involved in the interaction with vinculin.


Assuntos
Actinina/genética , Proteínas Aviárias/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Miopatias da Nemalina/patologia , Sarcômeros/metabolismo , Vinculina/metabolismo , Actinina/química , Actinina/metabolismo , Animais , Sítios de Ligação/genética , Células Cultivadas , Embrião de Galinha , Galinhas , Conectina , Deleção de Genes , Modelos Biológicos , Proteínas Musculares/metabolismo , Mutagênese Sítio-Dirigida , Miopatias da Nemalina/genética , Miopatias da Nemalina/metabolismo , Miopatias da Nemalina/veterinária , Ligação Proteica/genética , Domínios e Motivos de Interação entre Proteínas/genética , Mapeamento de Interação de Proteínas , Proteínas Quinases/metabolismo , Sarcômeros/genética , Sarcômeros/ultraestrutura , Espectrina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...